1
|
Zhang W, Ma F, Su X, Zhu M, Wang X. Antimicrobial peptide WK-13-3D promotes apoptosis, autophagy, and ubiquitination in triple-negative breast cancer via binding immunoglobulin protein (BiP). Chem Biol Interact 2025; 415:111530. [PMID: 40294882 DOI: 10.1016/j.cbi.2025.111530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/08/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE To elucidate the inhibitory mechanism of antimicrobial peptide WK-13-3D on triple-negative breast cancer (TNBC) by targeting the binding immunoglobulin protein (BiP), a key endoplasmic reticulum (ER) chaperone regulating unfolded protein response and tumor survival. METHODS TNBC cell lines (MDA-MB-231 and MDA-MB-468) were treated with WK-13-3D to assess proliferation, migration, invasion, and apoptosis. Pull-down assays identified interacting proteins, and Western blotting (WB) analyzed alterations in BiP, PERK, eIF2α, p-eIF2α, Caspase3, Cleaved-Caspase3, Bax, LC3, P62, AKT, p-AKT, mTOR, and p-mTOR. Transmission electron microscopy examined intracellular structures, while qPCR measured BiP mRNA levels. The effects of WK-13-3D on BiP ubiquitination were explored via co-immunoprecipitation (Co-IP). Animal tumor models were used to confirm the inhibitory effects, with BiP and Ki67 (a nuclear proliferation marker indicating actively dividing tumor cells) expression analyzed by immunohistochemistry (IHC). RESULTS WK-13-3D inhibited TNBC cell proliferation, migration, and invasion, while promoting apoptosis. Pull-down experiments identified 268 interacting proteins, with BiP being the most frequent. Databases (TIMER and TCGA) showed high BiP expression in breast cancer, associated with poor prognosis. WB assays revealed that WK-13-3D activated ER stress-induced apoptosis and autophagy via BiP. Co-IP demonstrated that WK-13-3D mediated BiP ubiquitination at sites 352 and 547 through K6 and K29 chains. IHC analysis further confirmed decreased Ki67 levels in WK-13-3D-treated tumors, reflecting suppressed proliferative activity. Animal experiments confirmed tumor growth inhibition. CONCLUSION WK-13-3D promotes apoptosis, autophagy and Ubiquitination in TNBC by modulating BiP.
Collapse
Affiliation(s)
- Wenjing Zhang
- College of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Fei Ma
- College of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Xuhong Su
- College of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Mingxing Zhu
- College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, PR China.
| | - Xiuqing Wang
- College of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, PR China; Ningxia Key Laboratory of Clinical Pathogenic Microorganisms, Ningxia Medical University, Yinchuan, 750004, PR China.
| |
Collapse
|
2
|
Xu X, Zhou H, Hong R, Gong J, Wan Y, Fu Q, Huang K, Li Y, Wang N, Zhao P, Cai K, Li F. A self-accelerating 'copper bomb' strategy activated innate and adaptive immune response against triple-negative breast cancer. Bioact Mater 2025; 49:193-206. [PMID: 40130080 PMCID: PMC11931225 DOI: 10.1016/j.bioactmat.2025.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 03/26/2025] Open
Abstract
Triple-negative breast cancer (TNBC) presents therapeutic challenges due to its aggressive, drug-resistance, and low immunological reactivity. Cuproptosis, an emerging therapeutic modality, is a promising strategic intervention for treating TNBC. Nonetheless, the effectiveness of cuproptosis is compromised by tumor adaptations, including the Warburg effect, increased intracellular glutathione (GSH), and copper efflux, thus breaking the barrier of cuproptosis is the basis for developing cuproptosis-based clinical therapies. Herein, a self-accelerating strategy utilizing a pH-responsive copper framework encapsulating glucose oxidase (GOx), modified with polyethylene glycol (PEG) and tumor-penetrating peptide (tLyp1) has been developed. Upon reaching the acidic tumor microenvironment, the released GOx increases intracellular acidity and hydrogen peroxide (H2O2). The elevated intracellular GSH and H2O2 serve as "fuel" to amplify the copper-based catalytic within tumor cells. Concurrently, the reduction of copper efflux proteins (ATP7B) and the depletion of GSH lead to copper overload in tumor cells, leading to cuproptosis via copper overload, mitochondrial disruption, and Fe-S protein instability. This constellation of interrelated events constitutes a potent "Copper Bomb," which concurrently triggers the immune system and effectively kills the tumor. It robustly engages innate and adaptive immunity via the release of mitochondrial DNA, facilitating the cGAS-STING pathway and precipitating immunogenic cell death. This process reverses the immunosuppressive tumor microenvironment, eliminates tumor cells, and suppresses metastasis, thus offering a novel therapeutic modality for the comprehensive treatment of triple-negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Xinzhi Xu
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Hang Zhou
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Ruixia Hong
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jiaqi Gong
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yujie Wan
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Qihuan Fu
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Kaifeng Huang
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Ying Li
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Na Wang
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Peng Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Fang Li
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
3
|
Jing Q, Zhang J, Yuan L, Zhang H, Lin W, Pei D, Di D, Yang L, Fan Z, Hai J. Copper-based hollow mesoporous nanogenerator with reactive oxygen species and reactive nitrogen species storm generation for self-augmented immunogenic cell death-mediated triple-negative breast cancer immunotherapy. J Colloid Interface Sci 2025; 688:688-702. [PMID: 40024101 DOI: 10.1016/j.jcis.2025.02.186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Although nanotheranostics have great potential in tumor immunotherapy, their effectiveness is often hindered by low immunogenic cell death (ICD) and inactivated immune responses in the tumor immunosuppressive microenvironment (TIME). Such vulnerability may lead to metastasis or recurrence, especially in triple-negative breast cancer (TNBC). Addressing this challenge, the study presents a multimodal immunotherapeutic approach using a self-enhanced ICD copper (Cu)-based hollow nanogenerator. This nanogenerator is activated by a near-infrared (NIR) laser to produce reactive oxygen species (ROS) and reactive nitrogen species (RNS) storms. Specifically, the nitric oxide (NO) donor l-Arginine (l-Arg) is loaded into hollow mesoporous Cu sulfide nanoparticles (HCuSNPs) with inherent NIR absorption and coated with tumor-targeting peptides (RGD), forming l-Arg@HCuSNPs-PEG-RGD (AHPR). In vitro and in vivo experiments demonstrate that AHPR can induce tumor thermal ablation, cuproptosis, and the generation of peroxynitrite anions (ONOO-) under NIR laser irradiation, resulting in multiple antitumor effects. Additionally, the nanogenerator enhances ICD through mechanisms such as mild-photothermal therapy (mPTT), cuproptosis, and ONOO- production, promoting immune cell infiltration and activation, and converting 'cold' tumors into 'hot' ones. By combining AHPR with the immune checkpoint inhibitor anti-programmed cell death protein ligand-1 antibody (αPD-L1), the study significantly improves the immunotherapy response rate in TNBC, offering a promising strategy to enhance TNBC immunotherapy efficacy.
Collapse
Affiliation(s)
- Quan Jing
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinlong Zhang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Longlong Yuan
- School of Pharmaceutical Sciences, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Haixia Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Wanquan Lin
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361002, China
| | - Dong Pei
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Duolong Di
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lichao Yang
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361002, China.
| | - Zhongxiong Fan
- School of Pharmaceutical Sciences, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China.
| | - Jun Hai
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China.
| |
Collapse
|
4
|
Diao B, Cai Y, Song D, Hu Y, Xie B, Kan Y, Hu X. A potential therapeutic molecule target: lncRNA AK023507 inhibits the metastasis of breast cancer by regulating the WNT/DOCK4/β-catenin axis. Breast Cancer Res Treat 2025; 211:727-741. [PMID: 40205246 DOI: 10.1007/s10549-025-07695-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 03/23/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE Breast cancer (BC) has become the most common malignant tumor in women worldwide. This study was carried out to find and validate a novel molecular therapeutic target for BC. METHODS Long non-coding RNA (lncRNA) AK023507 was selected as the study objects through microarray analysis. The function of lncRNA AK023507 was verified by various cell function experiments in vitro, subcutaneous tumorigenesis experiments, and lung metastasis model experiments in vivo. The RNA pull-down experiment and Western blot experiment were used to confirm the mechanism regulation pathway and the recovery experiment was used to verify it. TCGA datasets were used for clinical and immune function prediction analysis. RESULTS In vitro cell function tests and in vivo experiments suggested that overexpression of lncRNA AK023507 inhibited the proliferation and metastasis of BC cells. The RNA pull-down experiment and Western blot analysis validated that lncRNA AK023507 interacted with the dedicator of cytokinesis 4 (DOCK4) protein. Analysis of public databases predicted that DOCK4 is a potential prognostic risk factor associated with epithelial-mesenchymal transition (EMT) and central memory T cell (TCM) cellular immune infiltration. CONCLUSIONS LncRNA AK023507 inhibits the proliferation and metastasis of BC by regulating the DOCK4/β-catenin axis. This discovery will provide new potential therapeutic targets for BC.
Collapse
Affiliation(s)
- Biyu Diao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China
| | - Yangjun Cai
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China
- Department of Thyroid and Breast Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Dandan Song
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China
| | - Yingying Hu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China
| | - Bojian Xie
- Department of Thyroid and Breast Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Yang Kan
- Department of Thyroid and Breast Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Xiaoqu Hu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, No. 96, Fuxue Lane, Lucheng District, Wenzhou, 325000, China.
| |
Collapse
|
5
|
Cao X, Ren X, Song Y, Sun Q, Mao F, Shen S, Chen C, Zhou Y. High Expression of Calreticulin Affected the Tumor Microenvironment and Correlated With Worse Prognosis in Patients With Triple-Negative Breast Cancer. J Immunother 2025; 48:173-182. [PMID: 40123257 PMCID: PMC12052058 DOI: 10.1097/cji.0000000000000553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
Calreticulin (CALR) preserves reticular homeostasis by maintaining correct protein folding within the endoplasmic reticulum. Immunogenic cell death (ICD) is a regulated form of cell death and could activate adaptive immune response. As one of the damage-associated molecular patterns during ICD process, surface-exposed CALR resulted in the activation of adaptive immune response. Here, we evaluated the expression patterns of CALR in a cohort of 231 untreated triple-negative breast cancer (TNBC) and determined correlations between CALR expression and clinicopathologic parameters, programmed cell death ligand 1 (PD-L1) expression in immune cells (ICs), and survival. In addition, we analyzed a TNBC data set from The Cancer Genome Atlas to explore the relationship between mRNA expression of CALR and clinicopathologic features, IC infiltration, and survival. Tissue microarray results showed that high CLAR was strongly correlated with advanced stage ( P = 0.022), shorter disease-free survival ( P = 0.008) and overall survival ( P = 0.002), and independently predicted prognosis in TNBC. Spearman analyses demonstrated that CALR negatively correlated with PD-L1 in ICs ( r = -0.198, P = 0.003). Patients with low CALR and high PD-L1 in ICs had the best disease-free survival ( P = 0.013) and overall survival ( P = 0.004) compared with other patients, especially the patients with high CALR and low PD-L1 in ICs. In the "The Cancer Genome Atlas" cohort, CALR mRNA expression in tumors was significantly higher than that in normal tissues ( P < 0.001). CALR expression was strongly and positively related to other ICD-related genes. These findings demonstrated that the expression of CALR could independently predict the prognosis in patients with TNBC, and it may play a potential synergistic role in treatments involving immunotherapy.
Collapse
Affiliation(s)
- Xi Cao
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinyu Ren
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yu Song
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Feng Mao
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Songjie Shen
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chang Chen
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yidong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Fan G, Zuo S, Wang Z, Zhang S, Liu L, Luo H, Xie Y, Zhang Y, Li D. Targeting of the IL-33/Wnt axis restricts breast cancer stemness and metastasis. Sci Rep 2025; 15:18172. [PMID: 40414980 DOI: 10.1038/s41598-025-03260-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Interleukin-33 (IL-33) plays multifaceted roles in tumor progression, but its autocrine regulation of breast cancer stemness and metastasis via the Wnt pathway remains unclear. Here, we investigated the IL-33/ST2 axis in breast cancer using CRISPR/Cas9, single-cell RNA sequencing, and murine models (orthotopic 4T1 and spontaneous MMTV-PyMT). Elevated IL-33 levels correlated with aggressive subtypes and poor prognosis. IL-33 overexpression enhanced proliferation, migration, and cancer stem cell (CSC) marker expression (CD44, ALDH1) in 4T1 and MDA-MB-231 cells, whereas ST2 knockdown via CRISPR or adeno-associated virus (AAV) attenuated tumor growth and metastasis in vivo, reducing CSC frequency. Mechanistically, IL-33 activated Wnt/β-catenin signaling to promote stemness, which was reversed by the Wnt inhibitor XAV-939. Single-cell analysis revealed that IL-33 overexpression skewed the immune microenvironment toward immunosuppression, while ST2 knockdown restored antitumor immunity. Our findings establish an IL-33-Wnt axis as a critical driver of breast cancer aggressiveness and propose AAV-mediated ST2 silencing as a novel therapeutic strategy. Targeting this axis may offer dual benefits by suppressing stemness and enhancing immune surveillance, warranting clinical exploration for advanced breast cancer.
Collapse
Affiliation(s)
- Guanglin Fan
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Shuting Zuo
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhen Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Siwei Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Liping Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Haoge Luo
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yingdong Xie
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yan Zhang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China.
| | - Dong Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
7
|
He L, He J, Jiang T, Gong R, Wan X, Duan M, Chen Z, Cheng Y. Inhibition of UCH-L1 enhances immunotherapy efficacy in triple-negative breast cancer by stabilizing PD-L1. Eur J Pharmacol 2025; 1000:177743. [PMID: 40389130 DOI: 10.1016/j.ejphar.2025.177743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Recent research indicates that programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors show promise in treating triple-negative breast cancer (TNBC), but their efficacy is lower than anticipated, especially when used alone. Therefore, enhancing the anti-tumor immune response strategy for TNBC is crucial. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), involved in tumor cell regulation and a potential therapeutic target, has an undefined role in TNBC immunotherapy. In this study, we explored the inverse correlation between UCH-L1 and PD-L1 in TNBC patient tissues. Through in vitro experiments, we found that UCH-L1 negatively regulates PD-L1 by stabilizing the E3 ubiquitin ligase ariadne-1 homolog (ARIH1), which promotes PD-L1 ubiquitination and degradation. Further analysis in Balb/c mice xenograft tumors showed that UCH-L1 correlates with GZMB+/CD8+ T cell infiltration in TNBC, suggesting potential synergistic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. Overall, in TNBC, UCH-L1 stabilizes ARIH1, leading to low PD-L1 expression, which may explain the limited effectiveness of immunotherapy in TNBC patients. Our mouse experiments showed improved therapeutic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. These findings offer a new avenue for immunotherapy in TNBC patients.
Collapse
Affiliation(s)
- Linhao He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Jiaying He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Rong Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Xiaoya Wan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Mingwu Duan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Zonglin Chen
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China; Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China; NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, 410008, China; Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, 410011, China.
| |
Collapse
|
8
|
Zhu B, Wan H, Ling Z, Jiang H, Pei J. Machine learning and single-cell analysis uncover distinctive characteristics of CD300LG within the TNBC immune microenvironment: experimental validation. Clin Exp Med 2025; 25:167. [PMID: 40382513 PMCID: PMC12085369 DOI: 10.1007/s10238-025-01690-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
Investigating the essential function of CD300LG within the tumor microenvironment in triple-negative breast cancer (TNBC). Transcriptomic and single-cell data from TNBC were systematically collected and integrated. Four machine learning algorithms were employed to identify distinct target genes in TNBC patients. Specifically, CIBERSORT and ssGSEA algorithms were utilized to elucidate immune infiltration patterns, whereas TIDE and TCGA algorithms predicted immune-related outcomes. Moreover, single-cell sequencing data were analyzed to investigate the function of CD300LG-positive cells within the tumor microenvironment. Finally, immunofluorescence staining confirmed the significance of CD300LG in tumor phenotyping. After machine learning screening and independent dataset validation, CD300LG was identified as a unique prognostic biomarker for triple-negative breast cancer. Enrichment analysis revealed that CD300LG expression is strongly linked to immune infiltration and inflammation-related pathways, especially those associated with the cell cycle. The presence of CD8+ T cells and M1-type macrophages was elevated in the CD300LG higher group, whereas the abundance of M2-type macrophage infiltration showed a significant decrease. Immunotherapy prediction models indicated that individuals with low CD300LG expression exhibited better responses to PD-1 therapy. Additionally, single-cell RNA sequencing and immunofluorescence analyses uncovered a robust association between CD300LG and genes involved in tumor invasion. CD300LG plays a pivotal role in the tumor microenvironment of TNBC and represents a promising therapeutic target.
Collapse
Affiliation(s)
- Baoxi Zhu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Thyroid and Breast Surgery, Anhui No.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Hong Wan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zichen Ling
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Han Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Pei
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
9
|
Gao X, Wang T, Liu C, Li Y, Zhang W, Zhang M, Yao Y, Gao C, Liu R, Sun C. The integrated single-cell analysis interpret the lactate metabolism-driven immune suppression in triple-negative breast cancer. Discov Oncol 2025; 16:784. [PMID: 40377730 PMCID: PMC12084458 DOI: 10.1007/s12672-025-02605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Individuals with triple-negative breast cancer (TNBC) exhibit elevated lactate levels, which offers a valuable lead for investigating the molecular mechanisms underlying the tumor microenvironment (TME) and identifying more efficacious treatments. METHODS TNBC samples were classified based on lactate-associated genes. A single-cell transcriptomic approach was employed to examine functional differences across cells with varying lactate metabolism. Immunohistochemistry was used to explore the relationship between lactate metabolism and the CXCL12/CXCR4 signaling axis. In addition, utilizing machine learning techniques, we constructed a prognostic model based on lactic acid phenotype genes. RESULTS Lactate-associated gene-based stratification revealed increased immune cell infiltration and immune checkpoint expression in Lactate Cluster 1. Elevated lactate metabolism scores were observed in both cancer-associated fibroblasts (CAFs) and malignant cells. CAFs with high lactate metabolism exhibited immune suppression through the CXCL12/CXCR4 axis. Immunohistochemistry confirmed elevated LDHA, LDHB, CXCL12, and CXCR4 levels in the high lactate group. CONCLUSION This study elucidates the complex interplay between lactate and immune cells in TNBC and highlights the CXCL12/CXCR4 axis as a key pathway through which lactate mediates immune suppression, offering new insights into metabolic regulation within the TME. Furthermore, we developed a prognostic model based on lactate metabolism phenotype genes to predict the prognosis of TNBC patients and guide immunotherapy.
Collapse
Affiliation(s)
- Xinhai Gao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, 999078, Macao, China
| | - Tianhua Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, 999078, Macao, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Shandong Second Medical University, 261000, Weifang, Shandong, China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, 999078, Macao, China
| | - Wenfeng Zhang
- College of Traditional Chinese Medicine, Shandong Second Medical University, 261000, Weifang, Shandong, China
| | - Minpu Zhang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, 999078, Macao, China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, Shandong, China
| | - Chundi Gao
- College of Traditional Chinese Medicine, Shandong Second Medical University, 261000, Weifang, Shandong, China
| | - Ruijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, Shandong, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, 261000, Weifang, Shandong, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, Shandong, China.
| |
Collapse
|
10
|
Han Y, Hu M, Wang Y, Xu S, Jiang F, Wang Y, Liu Z. A coagulation-related long non-coding RNA signature to predict prognosis and immune features of breast cancer. Discov Oncol 2025; 16:662. [PMID: 40317354 PMCID: PMC12049355 DOI: 10.1007/s12672-025-02316-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/04/2025] [Indexed: 05/07/2025] Open
Abstract
Breast cancer (BC) remains one of the most common malignancies among women worldwide, with persistently poor prognosis despite advancements in diagnostics and therapies. Long non-coding RNAs (lncRNAs) and coagulation-related genes (CRGs) are increasingly recognized for their roles in prognosis and immune modulation. Using transcriptomic data from 1,045 BC patients in TCGA, we identified CRG-associated lncRNAs via coexpression analysis (Pearson |R|> 0.4, p < 0.001) and constructed a prognostic model through univariate Cox analysis, LASSO regression with tenfold cross-validation (λ = 0.05), and multivariate Cox analysis. The model stratified patients into high- and low-risk groups with distinct overall survival (HR = 3.21, p < 0.001) and demonstrated robust predictive accuracy (AUC = 0.795 at 1 year). Functional enrichment revealed immune-related pathways (e.g., cytokine signaling, PD-L1 regulation), and high-risk patients exhibited elevated tumor mutational burden (TMB) and PD-L1 expression, suggesting enhanced immunotherapy responsiveness. Drug sensitivity analysis identified 5 targeted agents (e.g., BIBW2992) with differential efficacy between risk groups. This CRG-lncRNA signature provides a novel tool for prognosis prediction and personalized immunotherapy in BC, illuminating crosstalk between coagulation and immune pathways.
Collapse
Affiliation(s)
- Yetao Han
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Mengsi Hu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yanzhong Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Shoufang Xu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Feiyu Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yingjian Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Zhiwei Liu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China.
| |
Collapse
|
11
|
Coleman C, Selvakumar T, Thurlapati A, Graf K, Pavuluri S, Mehrotra S, Sahin O, Sivapiragasam A. Harnessing Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Opportunities and Barriers to Clinical Integration. Int J Mol Sci 2025; 26:4292. [PMID: 40362529 PMCID: PMC12072607 DOI: 10.3390/ijms26094292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Triple-negative breast cancer (TNBC) continues to present a therapeutic challenge due to the fact that by definition, these cancer cells lack the expression of targetable receptors. Current treatment options include cytotoxic chemotherapy, antibody-drug conjugates (ADC), and the PD-1 checkpoint inhibitor, pembrolizumab. Due to high rates of recurrence, current guidelines for early-stage TNBC recommend either multi-agent chemotherapy or chemo-immunotherapy in all patients other than those with node-negative tumors < 0.5 cm. This approach can lead to significant long-term effects for TNBC survivors, driving a growing interest in de-escalating therapy where appropriate. Tumor infiltrating lymphocytes (TILs) represent a promising prognostic and predictive biomarker for TNBC. These diverse immune cells are present in the tumor microenvironment and within the tumor itself, and multiple retrospective studies have demonstrated that a higher number of TILs in early-stage TNBC portends a favorable prognosis. Research has also explored the potential of TIL scores to predict the response to immunotherapy. However, several barriers to the widespread use of TILs in clinical practice remain, including logistical and technical challenges with the scoring of TILs and lack of prospective trials to validate the trends seen in retrospective studies. This review will present the current understanding of the role of TILs in TNBC and discuss the future directions of TIL research.
Collapse
Affiliation(s)
- Cara Coleman
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Tharakeswari Selvakumar
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Aswani Thurlapati
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Kevin Graf
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Sushma Pavuluri
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Abirami Sivapiragasam
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| |
Collapse
|
12
|
Ding F, Zhao J, Wu X, Liu X, Yang Y, Li Y, Qiao L, Zhang Y. Impact of postmastectomy radiotherapy on locoregional recurrence and survival in patients with ypN0 breast cancer after neoadjuvant chemotherapy: A comprehensive meta-analysis and systematic review. J Cancer Res Ther 2025; 21:477-482. [PMID: 40317154 DOI: 10.4103/jcrt.jcrt_1279_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 02/18/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND The necessity of postmastectomy radiotherapy (PMRT) following neoadjuvant chemotherapy (NAC) in patients with ypN0 breast cancer remains a controversial clinical issue. To elucidate this issue, the present study performed a systematic review and meta-analysis, focusing on studies of clinically lymph node-negative patients exhibiting a favorable pathologic response to NAC. MATERIALS AND METHODS After a comprehensive literature search of PubMed and major oncology congress abstracts, 16 studies that met the inclusion criteria were included. A fixed or random effects model was used to assess the impact of PMRT on the local recurrence rate (LRR) and survival outcomes, where appropriate. RESULTS The pooled data from ten studies indicated that PMRT significantly reduced the LRR in patients with ypN0 breast cancer (risk ratio [RR], 0.52; 95% confidence interval [CI], 0.41-0.67, P < 0.00001). Moreover, a marginal increase in the overall survival (OS) rates was observed in nine studies (RR, 1.01; 95% CI, 1.00-1.02, P = 0.006), whereas no significant effect on disease-free survival (DFS) was found in six studies (RR, 1.01; 95% CI, 0.95-1.08, P = 0.72). CONCLUSIONS Our findings suggest that PMRT is associated with reduced LRR and slightly improved OS in patients with ypN0 breast cancer. However, prospective studies are needed to provide more evidence and inform clinical decision-making.
Collapse
Affiliation(s)
- Fangjie Ding
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, Shandong, China
| | - Junfeng Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xue Wu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, Shandong, China
| | - Xiaoman Liu
- Department of Oncology, BinZhou Medical University Affiliated Hospital, BinZhou Medical University, Binzhou, Shandong, China
| | - Yunxing Yang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, Shandong, China
| | - Ying Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lili Qiao
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, Shandong, China
| | - Yingying Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, Shandong, China
| |
Collapse
|
13
|
Zhang Y, Yang H, Jiang Y, Jiang Y, Mao R. Angiogenesis and immune microenvironment in triple-negative breast cancer: Targeted therapy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167880. [PMID: 40316057 DOI: 10.1016/j.bbadis.2025.167880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype that typically lacks effective targeted therapies, leading to limited treatment options. Chemotherapy remains the primary treatment modality; however, in recent years, new immunotherapy approaches, such as immune checkpoint inhibitors, have shown positive results in some patients. Although the development of TNBC is closely associated with BRCA gene mutations, the tumor immune microenvironment (TIME) plays a crucial role in tumor progression and immune escape. Tumor angiogenesis, the accumulation of immunosuppressive cells, and alterations in immune molecules collectively shape an environment unfavorable for anti-tumor immune responses. Tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) promote immune escape by secreting immunosuppressive factors. Therefore, combination strategies of anti-angiogenic and immune checkpoint inhibitory therapies have shown synergistic effects in clinical trials, while new targeted therapies such as TGF-β inhibitors and IL-1β inhibitors offer new options for TNBC treatment. With the development of personalized medicine, combining immunotherapy and targeted therapies brings new hope for TNBC patients.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Hao Yang
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Yanhong Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China..
| |
Collapse
|
14
|
Zhang Y, Yu M. Unmeasured Health Disparities: Hidden Confounders in Survival Outcomes. Clin Breast Cancer 2025:S1526-8209(25)00104-1. [PMID: 40414761 DOI: 10.1016/j.clbc.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Accepted: 04/25/2025] [Indexed: 05/27/2025]
Affiliation(s)
- Yining Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Mingtao Yu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Shao Y, Zhang Y, Chen J, Yang L, Wu M, Fan Z, Zhuang Z. Comprehensive analysis of breast cancer oxidative stress related gene signature: a combination of bulk and single-cell RNA sequencing analysis. Mamm Genome 2025:10.1007/s00335-025-10130-2. [PMID: 40274661 DOI: 10.1007/s00335-025-10130-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
Oxidative stress influences the tumor microenvironment, driving breast cancer progression and drug resistance. This study aimed to develop a prognostic gene signature based on oxidative stress-related genes (OSRGs) to assess patient outcomes and immune status. UCSC Xena ( http://xena.ucsc.edu/ ) and GEO ( https://www.ncbi.nlm.nih.gov/geo/ ) databases were used to obtain RNA-seq data and corresponding clinical information. The classification of OSRG subtypes was performed using consensus cluster. The oxidative stress related scoring (OSRS) model was established combining Lasso regression and multivariable Cox regression. The analysis of tumor mutation burden (TMB) and somatic mutation were carried out using the R package 'maftools'. Python package 'pySCENIC' was used to construct and analyze the transcription factor network. Additionally, immune infiltration was analyzed using R packages 'CIBERSORT' and 'ESTIMATE'. Three OSRG subgroups were identified and the Differentially Expressed Genes (DEGs) among them were enriched in humoral immunity, cytokine communication and drug metabolism pathways. OSRS model was established based on the DEGs and revealed association with patients' overall survival, somatic mutations, immune statuses, and drug resistance. Finally, transcription factor TFAP2B was identified as a key regulatory factor in high OSRS cells, and associated with a negative prognostic outcome in Basal-like breast cancer patients.
Collapse
Affiliation(s)
- Yuheng Shao
- Department of Radiation Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yumeng Zhang
- Department of Radiation Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jie Chen
- Department of Radiation Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Liang Yang
- Department of Radiation Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Meihong Wu
- Department of Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Zhiyuan Fan
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Zhigang Zhuang
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
16
|
Dai X, Cao B, Liu X, Meng W, Qiu Y, Sun Y, Zhang L, Li N, Liu Z, Li D, Xiao L, Li B, Zhang Q. Tumor vascular normalization by B7-H3 blockade augments T lymphocyte-mediated antitumor immunity. Eur J Pharmacol 2025; 993:177334. [PMID: 39892447 DOI: 10.1016/j.ejphar.2025.177334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/09/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Triple-negative breast cancer (TNBC), defined by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), presents unique clinical challenges and generally predicts a less favorable prognosis. Despite recent advancements in TNBC treatment, a subset of patients remains resistant to immunotherapy. B7-H3, a member of the B7 family of immune checkpoints, is correlated with poor outcomes in various cancers and is distinctively expressed in tumor vasculature, marking it as a potential biomarker for tumor-associated endothelial cells. We found high expression of B7-H3 in the endothelial cells of the postoperative tissue of TNBC patients. Elevated gene expression of CD276 (encoding B7-H3) and PECAM1 (encoding CD31) in TNBC is associated with poor prognosis. Anti-B7-H3 blockade reduces tumor burden and promotes lymphocyte infiltration in a TNBC mouse model. Additionally, anti-B7-H3 blockade promotes tumor vessel normalization and enhances programmed cell death ligand 1 (PD-L1) expression. Synergistic effects were observed when B7-H3 blockade was combined with programmed cell death protein 1 (PD-1) inhibition in the TNBC mouse model. Furthermore, anti-B7-H3 inhibits human umbilical vein endothelial cell (HUVEC) proliferation by suppression of the nuclear factor kappa-B (NF-κB) signaling pathway. Downregulation of B7-H3 expression in HUVECs promotes lymphocyte trans-endothelial migration. These findings suggest that B7-H3 represents a promising therapeutic target for TNBC, and the combination of anti-B7-H3 and anti-PD-1 therapies may have synergetic effects in treating TNBC.
Collapse
Affiliation(s)
- Xin Dai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China; Department of Oncology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Boran Cao
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinnan Liu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wangyang Meng
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiran Qiu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University School of Medicine, Shanghai, China
| | - Yidan Sun
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulu Zhang
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nan Li
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenyu Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dan Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lianbo Xiao
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Bin Li
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
17
|
Khaleel AQ, Altalbawy FMA, Jabir MS, F Hasan T, Jain V, Abbot V, Nakash P, Kumar MR, Mustafa YF, Jawad MA. CXCR4/CXCL12 blockade therapy; a new horizon in TNBC therapy. Med Oncol 2025; 42:161. [PMID: 40216617 DOI: 10.1007/s12032-025-02705-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/29/2025] [Indexed: 05/03/2025]
Abstract
The only subtype of breast cancer (BC) without specific therapy is triple-negative breast cancer (TNBC), which represents 15-20% of incidence cases of BC. TNBC encompasses transformed and nonmalignant cells, including cancer-associated fibroblasts (CAF), endothelial vasculature, and tumor-infiltrating cells. These nonmalignant cells, soluble factors (e.g., cytokines), and the extracellular matrix (ECM) form the tumor microenvironment (TME). The TME is made up of these nonmalignant cells, ECM, and soluble components, including cytokines. Direct cell-to-cell contact and soluble substances like cytokines (e.g., chemokines) may facilitate interaction between cancer cells and the surrounding TME. Through growth-promoting cytokines, TME not only enables the development of cancer but also confers therapy resistance. New treatment targets will probably be suggested by comprehending the processes behind tumor development and progression as well as the functions of chemokines in TNBC. In this light, several investigations have shown the pivotal function of the C-X-C motif chemokine ligand 12 (CXCL12 or SDF-1) axis and chemokine receptor type 4 (CXCR4) in the pathophysiology of TNBC. This review provides an overview of the CXCR4/CXCL12 axis' function in TNBC development, metastasis, angiogenesis, and treatment resistance. A synopsis of current literature on targeting the CXCR4/CXCL12 axis for treating and managing TNBC has also been provided.
Collapse
Affiliation(s)
- Abdulrahman Qais Khaleel
- Department of Medical Instruments Engineering, Al-Maarif University College, Al Anbar, 31001, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Thikra F Hasan
- College of Health&Medical Technology, Uruk University, Baghdad, Iraq
| | - Vicky Jain
- Department of Chemistry, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Vikrant Abbot
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Prashant Nakash
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | |
Collapse
|
18
|
Zhou J, Cheng A, Guo J, Liu Y, Li X, Chen M, Hu D, Wu J. Targeting PRDX2 to inhibit tumor growth and metastasis in triple-negative breast cancer: the role of FN1 and the PI3K/AKT/SP1 pathway. J Transl Med 2025; 23:434. [PMID: 40217291 PMCID: PMC11992733 DOI: 10.1186/s12967-025-06441-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is characterized by high invasiveness, high potential for metastasis, easy recurrence, and poor prognosis. There is an urgent need to develop new clinical treatments. METHODS This study utilized TNBC tissue microarrays to detect Peroxiredoxin 2 (PRDX2) expression levels and analyzed the correlation between PRDX2 and tumor invasion as well as invasion-related gene expression through the TCGA database. A stable PRDX2-knockdown triple-negative breast cancer cell line was established using lentiviral technology. The effects of PRDX2 on triple-negative breast cancer cell migration, invasion, and epithelial-mesenchymal transition (EMT) were investigated via wound healing assays, Transwell assays, qPCR, and Western blotting. RNA sequencing (RNA-seq), Western blotting, and dual luciferase reporter assays were performed to confirm that PRDX2 regulates FN1 expression through SP1. Furthermore, subcutaneous tumor xenograft models in nude mice were constructed to assess the effects of PRDX2 knockdown and the PRDX2 inhibitor Conoidin A on tumor growth in vivo. RESULTS Tissue microarray detection and correlative analysis revealed that PRDX2 is significantly upregulated in triple-negative breast cancer (TNBC) tumor tissues and positively correlated with genes associated with cell migration and invasion. Functional experiments demonstrated that in vitro knockdown of PRDX2 suppresses migration, invasion, and epithelial-mesenchymal transition (EMT) in TNBC cells. Furthermore, in vivo knockdown of PRDX2 or treatment with the PRDX2 inhibitor Conoidin A effectively reduced tumor burden. Mechanistic investigations utilizing RNA sequencing (RNA-seq) identified FN1 as a critical gene promoting TNBC cell migration and invasion. PRDX2 facilitates TNBC progression by activating the PI3K/AKT signaling pathway, which enhances SP1 binding to the FN1 gene promoter. This regulatory cascade ultimately drives tumor advancement in TNBC. CONCLUSIONS This study elucidates the role of the PRDX2/SP1/FN1 axis in TNBC migration and invasion, and highlights PRDX2 as a promising therapeutic target for triple-negative breast cancer.
Collapse
Affiliation(s)
- Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
- Department of Laboratory Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 232001, China
| | - Anqi Cheng
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Xuan Li
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Maoqian Chen
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China.
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China.
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, 232000, China.
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, 232000, China.
- Department of Laboratory Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 232001, China.
- , 168 Taifeng Street, Huainan, 232001, China.
| | - Jing Wu
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China.
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, 232000, China.
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, 232000, China.
- Department of Laboratory Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 232001, China.
| |
Collapse
|
19
|
Zhang Z, Liang S, Zheng D, Wang S, Zhou J, Wang Z, Huang Y, Chang C, Wang Y, Guo Y, Zhou S. Using Cancer-Associated Fibroblasts as a Shear-Wave Elastography Imaging Biomarker to Predict Anti-PD-1 Efficacy of Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:3525. [PMID: 40332007 PMCID: PMC12027048 DOI: 10.3390/ijms26083525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
In the clinical setting, the efficacy of single-agent immune checkpoint inhibitors (ICIs) in triple-negative breast cancer (TNBC) remains suboptimal. Therefore, there is a pressing need to develop predictive biomarkers to identify non-responders. Considering that cancer-associated fibroblasts (CAFs) represent an integral component of the tumor microenvironment that affects the stiffness of solid tumors on shear-wave elastography (SWE) imaging, wound healing CAFs (WH CAFs) were identified in highly heterogeneous TNBC. This subtype highly expressed vitronectin (VTN) and constituted the majority of CAFs. Moreover, WH CAFs were negatively correlated with CD8+ T cell infiltration levels and influenced tumor proliferation in the Eo771 mouse model. Furthermore, multi-omics analysis validated its role in immunosuppression. In order to non-invasively classify patients as responders or non-responders to ICI monotherapy, a deep learning model was constructed to classify the level of WH CAFs based on SWE imaging. As anticipated, this model effectively distinguished the level of WH CAFs in tumors. Based on the classification of the level of WH CAFs, while tumors with a high level of WH CAFs were found to exhibit a poor response to anti programmed cell death protein 1 (PD-1) monotherapy, they were responsive to the combination of anti-PD-1 and erdafitinib, a selective fibroblast growth factor receptor (FGFR) inhibitor. Overall, these findings establish a reference for a novel non-invasive method for predicting ICI efficacy to guide the selection of TNBC patients for precision treatment in clinical settings.
Collapse
Affiliation(s)
- Zhiming Zhang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shuyu Liang
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Dongdong Zheng
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shiyu Wang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jin Zhou
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ziqi Wang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yunxia Huang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Cai Chang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuanyuan Wang
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Yi Guo
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Shichong Zhou
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
20
|
Xu Y, Ge M, Xu Y, Yin K. Ferroptosis: a novel perspective on tumor immunotherapy. Front Immunol 2025; 16:1524711. [PMID: 40260246 PMCID: PMC12009862 DOI: 10.3389/fimmu.2025.1524711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025] Open
Abstract
Ferroptosis is a novel form of programmed cell death characterized by iron-dependent accumulation of reactive oxygen species (ROS) and lipid peroxidation. The execution of ferroptosis is intricately linked to both iron and lipid metabolism. Intriguingly, iron and lipid metabolism are also pivotal for maintaining the physiological function of immune cells. Research has revealed that ferroptosis can potentiate the immunogenicity of tumor cells and engage in intricate interactions with immune cells. Certain ferroptosis inducers have the capacity to augment the efficacy of immunotherapy by modulating the tumor immune microenvironment. Ferroptosis holds immense potential in cancer immunotherapy and is anticipated to emerge as a novel therapeutic target in the future landscape of cancer treatment. In this review, we primarily delineate the ferroptosis signaling pathways and metabolic processes pertinent to immune cells, and further summarize the roles of ferroptosis in tumor-infiltrating immune cells. Ultimately, we anticipate further elucidation of the mechanisms of ferroptosis in immunotherapy and envision that strategies targeting ferroptosis and immunotherapy will be expeditiously applied in clinical oncology practice.
Collapse
Affiliation(s)
| | | | | | - Kai Yin
- Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
21
|
Wang G, Wang S, Song W, Lu C, Chen Z, He L, Wang X, Wang Y, Shi C, Liu Z, Yu Y, Wang X, Tian Y, Li Y. Integrating multi-omics data reveals the antitumor role and clinical benefits of gamma-delta T cells in triple-negative breast cancer. BMC Cancer 2025; 25:623. [PMID: 40197136 PMCID: PMC11974128 DOI: 10.1186/s12885-025-14029-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Gamma-delta (γδ) T cells are a critical component of the tumor microenvironment and have been recognized as a promising biomarker and target for cancer therapy. Increasing evidence suggests that γδT cells play distinct roles in different cancers. However, the impact of γδT cells in breast cancer remains controversial. METHODS In this study, we investigated the role of γδT cells in breast cancer using a comprehensive approach, including bulk and single-cell sequencing, radiomics based on magnetic resonance imaging (MRI), genomic data, and immunohistochemistry. Single-cell RNA profiling was used to infer the potential lineage evolution of γδT cells and their interactions with other immune cells. Bulk RNA sequencing was included to uncover the heterogeneity in signaling pathways, as well as radiotherapy and immunotherapy responses, among patients with varying levels of γδT cell abundance. Genomic analysis was used to recognize the critical gene mutations with the infiltration of γδT cells. Immunohistochemistry was performed to validate the prognostic value of γδT cells in breast cancer patients. Lastly, radiomics was used to establish a correlation between the abundance of γδT cells and the features of MRI images. RESULTS The γδT cell infiltration was closely associated with favorable prognosis in triple-negative breast cancer (TNBC) but not in other subtypes of breast cancer. γδT cells may exert antitumor effects through intrinsic lineage evolution or interact with antigen-presenting cells through ligand-receptor pairs. Patients with a high γδT cell abundance may benefit more from chemotherapy or radiotherapy alone than their combination. Additionally, patients with a high γδT cell abundance were more likely to benefit from immunotherapy. Finally, we established a radiomic model based on dynamic contrast-enhanced-MRI, which indicated the potential for estimating the γδT cell abundance for patients with TNBC. CONCLUSION Our study provides novel insight and a theoretical basis for individualized therapy of patients with TNBC based on γδT cells.
Collapse
Affiliation(s)
- Guixin Wang
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Qixiangtai Road 22, He-Ping District, Tianjin, 300070, China
| | - Shuo Wang
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
| | - Wenbin Song
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Chenglu Lu
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
- Department of Pathology, Tangshan People's Hospital, Tangshan, Hebei, 063001, China
| | - Zhaohui Chen
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
| | - Long He
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Xiaoning Wang
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Yizeng Wang
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Cangchang Shi
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Zhaoyi Liu
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Yue Yu
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
| | - Xin Wang
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China.
| | - Yao Tian
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China.
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China.
| | - Yingxi Li
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Qixiangtai Road 22, He-Ping District, Tianjin, 300070, China.
| |
Collapse
|
22
|
Jia Y, Peng Z, Tian X, Guan Y, Han Y, Ji D, Lan B, Xu B, Fan Y. Single-cell sequencing exposes mast cell-derived CD52's anti-tumor action in breast cancer through the IL-6/JAK/STAT3 axis. Int J Biol Macromol 2025; 310:142879. [PMID: 40194575 DOI: 10.1016/j.ijbiomac.2025.142879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
The aggressive nature and rapid progression of triple-negative breast cancer (TNBC), coupled with a high likelihood of recurrence and mortality, underscore the critical need for effective treatments. While immunotherapy presents promising advantages for those with triple-negative breast cancer (TNBC), its efficacy is not universal. This disparity highlights the importance of investigating survival outcomes and prognostic factors for those TNBC patients who don't respond well to immunotherapy. Our study leverages both bulk and single-cell RNA sequencing data to conduct an in-depth analysis, revealing that genes associated with mast cells (PCMT1, VDAC1, YWHAB, BRD4, BTG1, and CD52) are pivotal in prognostication for TNBC patients. Laboratory experiments have further substantiated our findings, demonstrating that the overexpression of CD52 in mast cells impedes the proliferation, invasion, and metastasis of breast cancer cells. Further anti-CD52 treatment inhibiting breast tumor growth in vivo. Additionally, we have discovered that CD52 elicits its antitumor effects by meditating the IL-6/JAK/STAT3 signaling pathway. These insights not only enhance the prognostic significance of mast cells in TNBC but also pave the way for the development of novel targeted immunotherapy strategies.
Collapse
Affiliation(s)
- Yueran Jia
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zexi Peng
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xinzhu Tian
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ying Guan
- Department of Medical Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuhang Han
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dangyang Ji
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Lan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Ying Fan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
23
|
Wang Y, Jiang Y, Ding F, Lu J, Huang T, Zhong G, Zhu P, Ma Y, Li J, Wang X, Lin J, Zheng H, Wang W, Xu Y, Lyu X, Niu YS, Qi X, Li J, Chen B, He T, Zeng J, Ma Y. Phenotypes and cytokines of NK cells in triple-negative breast cancer resistant to checkpoint blockade immunotherapy. Breast Cancer Res 2025; 27:51. [PMID: 40181426 PMCID: PMC11969778 DOI: 10.1186/s13058-025-02003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/15/2025] [Indexed: 04/05/2025] Open
Abstract
Neoadjuvant checkpoint blockade immunotherapy (NATI) significantly prolonged outcomes for triple-negative breast cancer (TNBC). Residual tumor cells that survive NATI represent high-risk cell populations with metastatic potential and usually evade immunosurveillance by NK cells. Using an 82-protein panel, we here profiled single-cell membrane proteomics of CD56+ (NCAM1+) NK cells from tumor, peri-cancerous tissue, as well as peripheral blood from 28 TNBC patients post-NATI of residual cancer burden II/III. Unsupervised clustering resulted in several distinct clusters: 2 tumor-infiltrating NK (TINK) clusters with divergent functions of immune activation (TNFRSF7+) and suppression (SELL+); 2 immuno-suppressive peri-cancerous clusters; and 1 periphery-specific cluster. Considering the contradiction of the 2 TINK clusters, we further tested cytokine functions of SELL + and TNFRSF7 + TINKs by single-cell secreting proteomics using a 32-cytokine panel. Consistently, SELL + TINK clusters were characterized by immuno-suppressive secretion patterns (IL10+). A low proportion of SELL + TINK cluster and low proportion of IL10 + secreting SELL + TINK cluster (single-cell secreting proteomics) were both associated with better progression-free survival time. These findings were validated in an independent cohort of 15 patients during 16-month follow-up. Overall, we identified a distinct immuno-suppressive TINK cell group, featuring IL10 + secreting and SELL expression with a strong relation to poor survival prognosis in TNBC patients post-NATI.
Collapse
Affiliation(s)
- Youlong Wang
- Department of General Surgery, Hainan Hospital of People's Liberation Army General Hospital, Sanya, Hainan, China
| | - Yongluo Jiang
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Fadian Ding
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Institute of Abdominal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatobiliary and Pancreatic Surgery, National Regional Medical Center Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jun Lu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Tong Huang
- Department of General Surgery, General Hospital of XinJiang Military Command, Urumqi, XinJiang, China
| | - Guanqing Zhong
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Pengfei Zhu
- Department of Clinical Laboratory & Key Clinical Laboratory of Henan province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yue Ma
- Department of Laboratory Medicine, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jin Li
- Department of Anesthesiology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinjia Wang
- Department of Orthopedics and Spine Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Orthopedics and Spine Surgery, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jiacai Lin
- Department of Neurology, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan, China
| | | | - Weidong Wang
- Department of Orthopedics and Spine Surgery, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yiwei Xu
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiajie Lyu
- Internal Medicine Department, Jacobi Medical Center, 1400 Pelham Play S, Bronx, NY, USA
| | - Yu Si Niu
- Acute Communicable Disease Epidemiology Division, Dallas County Health and Human Services, Dallas, TX, USA
| | - Xin Qi
- Department of Gastroenterology, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan, China
| | - Jinjian Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Bocen Chen
- Key Laboratory of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, Hainan, China
| | - Tingting He
- Department of thoracic surgery, The First People's Hospital of Shangqiu City Affiliated to Xinxiang Medical University, Shangqiu, Henan, China
| | - Jiling Zeng
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yifei Ma
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
- Institute of Abdominal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
- Department of Hepatobiliary and Pancreatic Surgery, National Regional Medical Center Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
24
|
Shen J, Ye X, Hou H, Wang Y. Efficacy and Safety of Immunochemotherapy in Advanced Triple-negative Breast Cancer: A Meta-analysis of Randomised Clinical Trials. Clin Oncol (R Coll Radiol) 2025; 40:103783. [PMID: 39955967 DOI: 10.1016/j.clon.2025.103783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/19/2024] [Accepted: 01/30/2025] [Indexed: 02/18/2025]
Abstract
AIMS Based on the existing controversial clinical research results, this study conducted a comprehensive meta-analysis of relevant literature to clarify the benefits of immunochemotherapy (ICT)-which combines immune checkpoint inhibitors and chemotherapy (CT)-for patients with advanced triple-negative breast cancer (aTNBC). MATERILAS AND METHODS A thorough literature search was conducted up to February 15, 2024. Subsequently, meta-analyses were performed to aggregate hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS), odds ratios (ORs) for objective response rate (ORR) and relative risks (RRs) for adverse events (AEs). RESULTS Six randomised clinical trials (RCTs) involving 3,105 patients met the inclusion criteria. In comparison with CT, ICT yielded significant enhancements in PFS (HR, 0.80; 95%CI: 0.73-0.87), OS (HR, 0.87; 95%CI: 0.80-0.96), and ORR (OR, 1.34; 95%CI: 1.15-1.55) in the intention-to-treat population. However, ICT also exhibited an increase in grade ≥3 AEs (RR, 1.11; 95%CI: 1.04-1.19) and severe AEs (RR, 1.40; 95%CI: 1.18-1.66). Subgroup analyses revealed that ICT significantly improved PFS (HR, 0.67; 95%CI: 0.58-0.77), OS (HR, 0.75; 95%CI: 0.64-0.87), and ORR (OR, 1.47; 95%CI: 1.16-1.84) within the PD-L1-positive subgroup, whereas no statistically significant differences were detected for PD-L1-negative population. CONCLUSION ICT demonstrates superior efficacy over conventional CT in the treatment of aTNBC, albeit accompanied by heightened toxicity. Notably, the assessment of PD-L1 status may serve as a valuable biomarker in discerning aTNBC patients who are particularly predisposed to derive benefit from ICT. PROSPERO NUMBER CRD42024513270.
Collapse
Affiliation(s)
- J Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - X Ye
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - H Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| | - Y Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| |
Collapse
|
25
|
Fina E, Vitale E, De Summa S, Gadaleta-Caldarola G, Tommasi S, Massafra R, Brunetti O, Rizzo A. Liquid biopsy for guiding breast cancer immunotherapy. Immunotherapy 2025; 17:369-383. [PMID: 40083311 PMCID: PMC12045575 DOI: 10.1080/1750743x.2025.2479426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 03/16/2025] Open
Abstract
Liquid biopsy is a laboratory test used to detect and analyze circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and other tumor-derived components, in a blood sample. In the context of breast cancer (BC), liquid biopsies hold significant promise for guiding the use of immune checkpoint inhibitors and immune-based combinations, offering real-time insights into tumor dynamics, treatment response, and resistance mechanisms. This review explores the role of liquid biopsy in BC immunotherapy, focusing on its applications, benefits, issues, and current and future research directions.
Collapse
Affiliation(s)
- Emanuela Fina
- Thoracic Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II, Bari, Italy
| | - Simona De Summa
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | | | - Stefania Tommasi
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Raffaella Massafra
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II, Bari, Italy
| | - Oronzo Brunetti
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II, Bari, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II, Bari, Italy
| |
Collapse
|
26
|
Huang J, Shi J, Ma N, Li Y, Jin W, Zhang H, Zhang X, Luo N, Ding Y, Xie Q, Li Q, Xiong Y. Celastrol-loaded ginsenoside Rg3 liposomes enhance anti-programmed death ligand 1 immunotherapy by inducing immunogenic cell death in triple-negative breast cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156514. [PMID: 39986227 DOI: 10.1016/j.phymed.2025.156514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC), characterized by high heterogeneity and invasiveness. Currently, inducing immunogenic cell death (ICD) of tumor cells through approaches such as radiotherapy and chemotherapy is an effective strategy to enhance the response to anti-programmed death-ligand 1 antibody (aPD-L1) therapy in TNBC. However, radiotherapy and chemotherapy treatments often upregulate PD-L1 expression in tumor cells, thereby weakening the tumor cells' response to aPD-L1. Celastrol exhibits broad-spectrum and potent anti-tumor activity, efficiently inducing ICD without increasing PD-L1 levels in tumor cells. PURPOSE This study aims to elucidate the tumor-targeting effects of celastrol-loaded liposomes and its synergistic efficacy and mechanism of action in combination with aPD-L1 against TNBC. METHODS The Rg3 liposomes loaded with celastrol (Cel-Rg3-Lp) were prepared using the thin-film hydration method. BALB/c mice were utilized to establish an in situ breast cancer model. Mice were intravenously injected with Cel-Rg3-Lp at a dosage of celastrol 1 mg/kg once every two days for a total of 7 injections. Flow cytometry, western blot, and immunofluorescence techniques were employed to investigate the synergistic effects and mechanisms of Cel-Rg3-Lp combined with aPD-L1 in the treatment of TNBC. RESULTS The findings of this study demonstrate that after 7 administrations of Cel-Rg3-Lp (1 mg/kg celastrol, intravenously), significant anti-tumor effects are observed, including the recruitment of CD8+T cells and dendritic cells (DCs), while reducing the infiltration of immunosuppressive cells. The therapeutic efficacy was further enhanced when combined with aPD-L1. Additionally, Cel-Rg3-Lp markedly downregulated glucose-regulated protein 78 (GRP78) expression, thereby inducing ICD in tumor cells. CONCLUSION This study successfully constructed a multifunctional liposome and proposed a mechanism for inducing ICD through the GRP78-endoplasmic reticulum stress pathway. The liposome downregulates GRP78, triggering endoplasmic reticulum stress in tumor cells, inducing ICD, activating DCs, and enhancing antigen presentation to T cells. This improves the tumor immune microenvironment and provides a theoretical foundation for combining Cel-Rg3-Lp with aPD-L1 in the treatment of TNBC. This mechanism opens unique prospects for using celastrol in TNBC therapy and enhancing the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Jingyi Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jingbin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ninghui Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yujie Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wanyu Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Xin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ningchao Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ye Ding
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Qiong Xie
- Gynecology Department, Zhoushan Hospital of Traditional Chinese Medicine (Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University), Zhoushan, Zhejiang 316000, China.
| | - Qiushuang Li
- Center of Clinical Evaluation and Analysis, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310053, China.
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
27
|
Al-Mansour FSH, Almasoudi HH, Albarrati A. Mapping molecular landscapes in triple-negative breast cancer: insights from spatial transcriptomics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04057-3. [PMID: 40119898 DOI: 10.1007/s00210-025-04057-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
The tumor microenvironment (TME) of triple-negative breast cancer (TNBC) is a highly heterogeneous and very aggressive form of the disease that has few suitable treatment options; however, spatial transcriptomics (ST) is a powerful tool for elucidation of the TME in TNBC. Because of its spatial context preservation, ST has a unique capability to map tumor-stroma interactions, immune infiltration, and therapy resistance mechanisms (which are key to understanding TNBC progression), compared with conventional transcriptomics. This review shows the use of ST in TNBC, its utilization in spatial biomarker identification, intratumoral heterogeneity definition, molecular subtyping refinement, and prediction of immunotherapy responses. Recent insight from ST-driven insights has explained the key spatial patterns on immune evasion, chemotherapy resistance, racial disparities of TNBC, and aspects for patient stratification and therapeutic decision. With the integration of ST with the subjects of proteomics and imaging mass cytometry, this approach has been enlarged and is now applied in precision medicine and biomarker discovery. Recently, advancements in AI-based spatial analysis for tumor classification, identification of biomarkers, and creation of therapy prediction models have occurred. However, continued developments in ST technologies, computational tools, and partnerships amongst multiple centers to facilitate the integration of ST into clinical routine practice are needed to unlock novel therapeutic targets.
Collapse
Affiliation(s)
- Fares Saeed H Al-Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ali Albarrati
- Rehabilitation Sciences Department, College of Applied Medical Sciences, King Saud University, 11451, Riyadh, Saudi Arabia.
| |
Collapse
|
28
|
He J, Liu N, Zhao L. New progress in imaging diagnosis and immunotherapy of breast cancer. Front Immunol 2025; 16:1560257. [PMID: 40165974 PMCID: PMC11955504 DOI: 10.3389/fimmu.2025.1560257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Breast cancer (BC) is a predominant malignancy among women globally, with its etiology remaining largely elusive. Diagnosis primarily relies on invasive histopathological methods, which are often limited by sample representation and processing time. Consequently, non-invasive imaging techniques such as mammography, ultrasound, and Magnetic Resonance Imaging (MRI) are indispensable for BC screening, diagnosis, staging, and treatment monitoring. Recent advancements in imaging technologies and artificial intelligence-driven radiomics have enhanced precision medicine by enabling early detection, accurate molecular subtyping, and personalized therapeutic strategies. Despite reductions in mortality through traditional treatments, challenges like tumor heterogeneity and therapeutic resistance persist. Immunotherapies, particularly PD-1/PD-L1 inhibitors, have emerged as promising alternatives. This review explores recent developments in BC imaging diagnostics and immunotherapeutic approaches, aiming to inform clinical practices and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Jie He
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nan Liu
- Department of Translational Medicine and Clinical Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Zhao
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
29
|
Li Q, Ye Z, Wang G, Chen Y, Deng J, Wang D, Wang Y. Natural Products as Novel Therapeutic Agents for Triple-Negative Breast Cancer: Current Evidence, Mechanisms, Challenges, and Opportunities. Molecules 2025; 30:1201. [PMID: 40141978 PMCID: PMC11944566 DOI: 10.3390/molecules30061201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 03/28/2025] Open
Abstract
Breast cancer (BC) tops the list of causes for female fatalities globally, with the elusive triple-negative breast cancer (TNBC) constituting 10-20% of all cases. Current clinical strategies for combating TNBC encompass a multifaceted approach, including surgical intervention, radiation therapy, chemotherapy, and advanced targeted drugs and immunotherapies. While these modalities have catalyzed significant advancements in TNBC management, lingering limitations continue to pose formidable challenges. There is an acute need for novel therapeutics in the realm of TNBC treatment. Natural products (NPs) have emerged as a rich reservoir for pharmaceutical innovation, owing to their extraordinary range of structures and physicochemical properties. Scholars have reported diverse evidence of NPs' efficacy against TNBC. This review aims to comprehensively explore the bioactive constituents, specifics and commonalities of chemical structure, and pharmacological mechanisms of NPs, specifically examining their multifaceted roles in impeding TNBC. NPs, which have recently garnered significant interest, are intriguing in terms of their capacity to combat TNBC through multifaceted mechanisms, including the suppression of tumor cell proliferation, the induction of apoptosis, and the inhibition of tumor metastasis. These natural agents primarily encompass a range of compounds, including terpenoids, glycosides, phenolic compounds, and alkaloids. An in-depth exploration has unveiled their involvement in key signaling pathways, including the transforming growth factor-beta (TGF-β), vascular endothelial growth factor A (VEGFA), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT), Wingless/Int-1 (Wnt) /β-catenin, and mitogen-activated protein kinase (MAPK) pathways. Meanwhile, this review also looks at the challenges and opportunities that arise from harnessing natural compounds to influence TNBC, while outlining the prospective trajectory for future research in the field of NPs.
Collapse
Affiliation(s)
- Qingzhou Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Guilin Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Yuhui Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Jinghong Deng
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Dong Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| | - Yumei Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Z.Y.); (G.W.); (Y.C.); (J.D.)
| |
Collapse
|
30
|
Fan J, Qin Y, Qiu W, Liang J, Xiao C, Xie Q, Tong C, Yuan L, Long Y, Liu B. Gamabufotalin loaded micro-nanocomposites for multimodal therapy of metastatic TNBC by efficiently inducing ICD. Biomaterials 2025; 314:122851. [PMID: 39366186 DOI: 10.1016/j.biomaterials.2024.122851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/30/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Gamabufotalin (CS-6), a main active compound derived from Chinese medicine Chansu, exhibits a robust inhibitory effect on programmed death-ligand 1 (PD-L1) in triple-negative breast cancer (TNBC) cells. Despite its potential for tumor therapy, the medical application of CS-6 is constrained by its hydrophobic nature, lack of targeting capability, and weak immunogenic cell death (ICD) effect. To address these limitations and improve the therapeutic efficiency of this drug against metastatic TNBC, we designed a new kind of CS-6@CPB-S.lux that integrates carboxy-Prussian blue nanoparticles (CPB NPs), CS-6, and attenuated Salmonella typhimurium (S.lux) for TNBC therapy. In vitro and in vivo results have confirmed that CS-6@CPB NPs were efficiently delivered to neoplastic tissue by the tumor hypoxic chemotaxis property of S.lux, wherein the nanomedicine induced significant tumor cell necroptosis and apoptosis via photothermal therapy (PTT) of CPB NPs and chemotherapy of CS-6, which elicited ICD and inhibited PD-L1 expression, resulting in dendritic cells (DCs) maturation and effector T cells activation to comprehensively eliminate tumors. Additionally, the CS-6@CPB-S.lux + Laser treatment significantly transformed the immunosuppressive tumor microenvironment (TME), enhancing antitumor immunity through promoting the polarization of tumor-associated macrophages into antitumorigenic M1 and reducing Tregs recruitment. Consequently, this comprehensive therapy not only inhibited primary and abscopal tumor progression but also prevented TNBC metastasis, which significantly prolonged survival time in animal models. In summary, these findings indicated an alternative approach for metastatic TNBC therapy.
Collapse
Affiliation(s)
- Jialong Fan
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Yan Qin
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China; TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wensheng Qiu
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Jiahao Liang
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Chang Xiao
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Qian Xie
- Department of Pharmacy, Maternal and Child Health of Hunan Province, Changsha, 410008, China
| | - Chunyi Tong
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Liqin Yuan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Ying Long
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China.
| | - Bin Liu
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China; NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
31
|
Lu X, Gou Z, Chen H, Li L, Chen F, Bao C, Bu H, Zhang Z. Extracellular matrix cancer-associated fibroblasts promote stromal fibrosis and immune exclusion in triple-negative breast cancer. J Pathol 2025; 265:385-399. [PMID: 39846260 DOI: 10.1002/path.6395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/05/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
The impact of high heterogeneity of cancer-associated fibroblasts (CAFs) on triple-negative breast cancer (TNBC) immunotherapy response has not been fully elucidated, restricting progress in precision immuno-oncology. We integrated single-cell transcriptomic data from 18 TNBC patients and analyzed fibroblast subpopulations. Extracellular matrix CAFs (ecmCAFs) were identified as a fibroblast subpopulation with distinct ECM-associated characteristics. The ecmCAFs were significantly enriched in TNBC patients with residual disease after neoadjuvant immunotherapy and contributed to a fibrotic tumor microenvironment and T-cell exclusion. Secreted phosphoprotein 1 (SPP1) positive macrophages (SPP1+ Mφs) were closely localized to ecmCAFs and produced more transforming growth factor beta (TGFB1), interleukin 1 beta (IL1B), and SPP1 under hypoxic conditions. SPP1+ Mφs were found to facilitate the differentiation of normal breast fibroblasts to ecmCAFs, thus promoting ECM remodeling and stromal fibrosis. Our work revealed the critical role of ecmCAFs in generating a desmoplastic architecture and driving immunosuppression in TNBC. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xunxi Lu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, PR China
- Laboratory of Breast Pathology and Artificial Intelligence, West China Hospital, Sichuan University, Chengdu, PR China
| | - Zongchao Gou
- Breast Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, PR China
| | - Hong Chen
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Li Li
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Fei Chen
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Chunjuan Bao
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, PR China
- Laboratory of Breast Pathology and Artificial Intelligence, West China Hospital, Sichuan University, Chengdu, PR China
| | - Zhang Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, PR China
- Laboratory of Breast Pathology and Artificial Intelligence, West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
32
|
Wu F, Feng X, Gao W, Zeng L, Xu B, Chen Z, Zheng C, Hu X, Xu S, Song H, Zhou X, Liu Z. Engineering a Self-Delivery Nanoplatform for Chemo-Photodynamic-Immune Synergistic Therapies against Aggressive Melanoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11634-11652. [PMID: 39960055 DOI: 10.1021/acsami.4c18469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The effectiveness of immunotherapy in killing melanoma is hindered by a T-cell deficiency and the lack of tumor immunogenicity. Consequently, there is an urgent need for a platform that can further activate the immune system and boost the immune response of the host to tumors. Compared with monotherapy, combination therapy shows promise in improving treatment efficacy and response rates. This study introduces the pioneering use of a rationally designed active targeting nanoplatform to bind axitinib, paclitaxel, and verteporfin to human serum albumin (APV@HSA NPs). APV@HSA NPs have demonstrated the capability to induce dual-induced apoptosis in tumor cells through chemo- and photodynamic effects, while also enhancing immunogenic cell death and promoting dendritic cell maturation. Additionally, the platform promoted the production of CD8+ T cells and memory T cells and inhibited vascular endothelial growth factor via axitinib, facilitating the infiltration of immune effector cells and optimizing chemo-photodynamic immunotherapy. Hence, amplified chemo-photodynamic-immunological nanomedicines with excellent biocompatibility have been redesigned to inhibit the tumor microenvironment and combat the growth of primary tumor and lung metastasis. This approach initiates a series of immune responses, presenting a promising therapeutic strategy for melanoma.
Collapse
Affiliation(s)
- Fei Wu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Xianquan Feng
- Fujian Provincial Key Laboratory of Transplant Biology, Laboratory of Basic Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Wenhao Gao
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Lingjun Zeng
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Bingbing Xu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Zhenzhen Chen
- Department of Clinical Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, Fujian Province 350025, China
| | - Changqing Zheng
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Xiaomu Hu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Shiying Xu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Hongtao Song
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Xin Zhou
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| | - Zhihong Liu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou 350025, China
| |
Collapse
|
33
|
Yang J, Chen M, Li R, Sun Y, Ye P, Fang K, Wang C, Shi S, Dong C. A responsive cocktail nano-strategy breaking the immune excluded state enhances immunotherapy for triple negative breast cancer. NANOSCALE 2025; 17:4610-4623. [PMID: 39810651 DOI: 10.1039/d4nr03054k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The exclusion of immune cells from the tumor can limit the effectiveness of immunotherapy in triple negative breast cancer (TNBC). The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway plays a crucial role in priming adaptive anti-tumor immunity through the production of type I interferons (IFNs), facilitating the maturation of dendritic cells (DCs) and the function of T cells. Although the increased expression of programmed death-ligand 1 (PD-L1) upon STING activation is favorable for amplifying the efficacy of immune checkpoint inhibitors (ICIs) and realizing combination therapy, the penetration barrier remains a major obstacle. Herein, we fabricated a smart-responsive nanosystem (B&V@ZB-MCL) by integrating the extracellular matrix (ECM)-degrading drug losartan with a STING agonist (Vadimezan, abbreviated to Vad) and a PD-L1 inhibitor (BMS-1). Losartan was first released in the acidic tumor microenvironment to overcome the physical barrier, enhancing the penetration of immunotherapeutic components. Under the triggering of 1O2 generated by a photosensitizer (Ce6), the reactive oxygen species (ROS)-sensitive degradation of the nanocore ensured the site-directed release of Vad and BMS-1. The released Vad and damaged tumor DNA activated immune responses through the cGAS-STING pathway, while the elevated expression level of PD-L1 promoted the anti-tumor effect of BMS-1. Significant degradation of collagen fibers, restoration of immune effector cells, and lower tumor volume were observed in this integrated triple drug sequential therapy, which provides a promising prospect for TNBC treatment.
Collapse
Affiliation(s)
- Jingxian Yang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Mengyao Chen
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Ruihao Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Yanting Sun
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Pingting Ye
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Kang Fang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Chunhui Wang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Shuo Shi
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| |
Collapse
|
34
|
Sun W, Huang S, Sun Z, Zhao Q, Li J, Wen B, Liu J, Deng G, Sun C. Exosome Platform with Three-in-One Functionality of mRNA Therapy, Immune Checkpoint Blockade, and Mild Photothermal Therapy for the Treatment of Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10328-10341. [PMID: 39905637 DOI: 10.1021/acsami.4c17804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Immune checkpoint blockade (ICB) has shown promising potential for treating triple-negative breast cancer (TNBC), but its efficacy is limited, mainly due to the "cold," suppressive immune tumor microenvironment (TME). Therefore, improving the TME is crucial for enhancing therapeutic effects against TNBC. Here, we presented a multifunctional nanotherapeutic platform (ALEvs@LNPs) designed to combine cytokine-sensitized mild photothermal therapy, ICB, and interleukin-2 (IL2) mRNA therapy to reprogram the TME, thereby improving the efficacy of ICB therapy in TNBC. Tumor cell-derived exosome-camouflaged lipid nanoparticles with antilymphocyte activation gene-3 (LAG3) were developed to codeliver the photothermal agent IR806, IL2 mRNA, and LAG3 inhibitory antibody (anti-LAG3). Mild photothermal therapy facilitated the reprogramming of "cold" tumors into "hot," thereby enhancing the therapeutic effects of ICB. Meanwhile, ICB also promoted cytokine secretion, increasing the sensitivity of tumor cells to heat. Additionally, IL2 mRNA therapy induced T-cell proliferation and activation, further augmenting the efficacy of ICB. Together, these three therapies established a positive feedback loop that enhanced the therapeutic effects of ICB. This multifunctional nanotherapeutic platform effectively reprogrammed the "cold," suppressive immune TME, offering a promising strategy for TNBC treatment.
Collapse
Affiliation(s)
- Wenwen Sun
- Shandong Provincial Hospital, Shandong University, Jinan 250021, P. R. China
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Shiyun Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Zhihong Sun
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Qi Zhao
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Jie Li
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Baoyu Wen
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Jie Liu
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Guanjun Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Chengming Sun
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| |
Collapse
|
35
|
Huang L, Liu Y, Shi Y, Sun Q, Li H, Sun C. Comprehensive single-cell analysis of triple-negative breast cancer based on cDC1 immune-related genes: prognostic model construction and immunotherapy potential. Discov Oncol 2025; 16:206. [PMID: 39969635 PMCID: PMC11839968 DOI: 10.1007/s12672-025-01929-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Various components of the immunological milieu surrounding tumors have become a key focus in cancer immunotherapy research. There are currently no reliable biomarkers for triple-negative breast cancer (TNBC), leading to limited clinical benefits. However, some studies have indicated that patients with TNBC may achieve better outcomes after immunotherapy. Therefore, this study aimed to identify molecular features potentially associated with conventional type 1 dendritic cell (cDC1) immunity to provide new insights into TNBC prognostication and immunotherapy decision-making. METHODS Single-cell ribonucleic acid sequencing data from the Gene Expression Omnibus database were analyzed to determine which genes are differentially expressed genes (DEGs) in cDC1s. We then cross-referenced cDC1-related DEGs with gene sets linked to immunity from the ImmPort and InnateDB databases to screen for the genes linked to the immune response and cDC1s. We used univariate Cox and least absolute shrinkage and selection operator regression analyses to construct a risk assessment model based on four genes in patients with TNBC obtained from the Cancer Genome Atlas, which was validated in a testing group. This model was also used to assess immunotherapy responses among the IMvigor210 cohort. We subsequently utilized single sample Gene Set Enrichment Analysis, CIBERSORT, and ESTIMATE to analyze the immunological characteristics of the feature genes and their correlation with drug response. RESULTS We identified 93 DEGs related to the immune response and cDC1s, of which four (IDO1, HLA-DOB, CTSD, and IL3RA) were substantially linked to the overall survival rate of TNBC patients. The risk assessment model based on these genes stratified patients into high- and low-risk groups. Low-risk patients exhibited enriched ''hot tumor'' phenotypes, including higher infiltration of memory-activated CD4 + T cells, CD8 + T cells, gamma delta T cells, and M1 macrophages, as well as elevated immune checkpoint expression and tumor mutational burden, suggesting potential responsiveness to immunotherapy. Conversely, high-risk patients displayed "cold tumor" characteristics, with higher infiltration of M0 and M2 macrophages and lower immune scores, which may be poorer in response to immunotherapy. However, experimental validation and larger clinical studies are necessary to confirm these findings and explore the underlying mechanisms of the identified genes. CONCLUSION This study developed a robust risk assessment model using four genes that effectively forecast the outcome of patients with TNBC and have the potential to guide immunotherapy. This model provided new theoretical insights for knowing the TNBC immune microenvironment and developing personalized treatment strategies.
Collapse
Affiliation(s)
- Linan Huang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China
| | - Yiran Liu
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China
| | - Yulin Shi
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Qi Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China.
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| |
Collapse
|
36
|
Cai WY, Cai XX, Fei YR, Ye R, Song DM, Hu D, Zhang WW, Xia MF, Yang XX. DNA methylation and immune evasion in triple-negative breast cancer: challenges and therapeutic opportunities. Front Oncol 2025; 15:1534055. [PMID: 39980537 PMCID: PMC11839428 DOI: 10.3389/fonc.2025.1534055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Chemotherapy remains the primary treatment option, yet TNBC frequently develops resistance, leading to relapse and metastasis. Emerging evidence highlights the potential of combining DNA methylation inhibitors with immune checkpoint inhibitors (ICIs). DNA methylation contributes to immune escape by silencing immune-regulatory genes, thereby reducing the tumor's visibility to immune cells. Reversing this epigenetic modification can reinvigorate immune surveillance and enhance the efficacy of immunotherapies. This review discusses the role of DNA methylation in TNBC progression and immune evasion, focusing on recent advances in combination therapies involving DNA methylation inhibitors and ICIs. We discuss the underlying mechanisms that enable these therapeutic synergies, preclinical and clinical evidence supporting the approach, and the challenges posed by tumor heterogeneity, drug resistance, and toxicity. Finally, we explore the potential for personalized treatment strategies incorporating multi-omics data to optimize therapeutic outcomes. The integration of epigenetic therapies and immunotherapy offers a promising avenue for improving survival in TNBC patients.
Collapse
Affiliation(s)
- Wen-yu Cai
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xin-xian Cai
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi-ran Fei
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rui Ye
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ding-ming Song
- Department of Urology, Jinzhou Medical University, The First Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Dan Hu
- Department of Clinical Lab, The Cixi Integrated Traditional Chinese and Western Medicine Medical and Health Group Cixi Red Cross Hospital, Cixi, China
| | - Wan-wan Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Ming-fei Xia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xiao-xiao Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
37
|
Wu Y, Liu Y, Wu H, Tong M, Du L, Ren S, Che Y. Advances in Ultrasound-Targeted Microbubble Destruction (UTMD) for Breast Cancer Therapy. Int J Nanomedicine 2025; 20:1425-1442. [PMID: 39925678 PMCID: PMC11804227 DOI: 10.2147/ijn.s504363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
Breast cancer is one of the most common types of cancer in women worldwide and is a leading cause of cancer deaths among women. As a result, various treatments have been developed to combat this disease. Breast cancer treatment varies based on its stage and type of pathology. Among the therapeutic options, ultrasound has been employed to assist in the treatment of breast cancer, including radiation therapy, chemotherapy, targeted immunotherapy, hormonal therapy, and, more recently, radiofrequency ablation for early-stage and inoperable patients. One notable advancement is ultrasound-targeted microbubble destruction (UTMD), which is gradually becoming a highly effective and non-invasive anti-tumor modality. This technique can enhance chemical, genetic, immune, and anti-vascular therapies through its physical and biological effects. Specifically, UTMD improves drug transfer efficiency and destroys tumor neovascularization while reducing toxic side effects on the body during tumor treatment. Given these developments, the application of ultrasound-assisted therapy to breast cancer has gained significant attention from research scholars. In this review, we will discuss the development of various therapeutic modalities for breast cancer and, importantly, highlight the application of ultrasound microbubble-targeted disruption techniques in breast cancer treatment.
Collapse
Affiliation(s)
- Yunfeng Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Yuxi Liu
- Department of Ultrasound, Shandong Second Medical University Affiliated Hospital, Shan Dong, Weifang, People’s Republic of China
| | - Han Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Mengying Tong
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Linyao Du
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Shuangsong Ren
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Ying Che
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| |
Collapse
|
38
|
Wang H, Cao Y, Zhang L, Zhao Q, Li S, Li D. RBM15 Drives Breast Cancer Cell Progression and Immune Escape via m6A-Dependent Stabilization of KPNA2 mRNA. Clin Breast Cancer 2025; 25:96-107. [PMID: 39488447 DOI: 10.1016/j.clbc.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/02/2024] [Accepted: 09/16/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Breast cancer is the most frequently diagnosed cancer among women worldwide with high morbidity and mortality. Previous studies have indicated that RNA-binding motif protein-15 (RBM15), an N6-methyladenosine (m6A) writer, is implicated in the growth of breast cancer cells. Herein, we aimed to explore the function and detailed mechanism of RBM15 in breast cancer. METHODS In this research, UALCAN databases were applied to analyze the expression of RBM15 or Karyopherin-2 alpha (KPNA2) in BRCA. RBM15 and KPNA2 mRNA levels were determined using real-time quantitative polymerase chain reaction (RT-qPCR) assay. RBM15, KPNA2, and Programmed cell death ligand 1 (PD-L1) protein levels were measured using western blot. Cell proliferation, migration, and invasion were assessed using 5-ethynyl-2'-deoxyuridine (EdU) and Transwell assays. The biological role of RBM15 on breast cancer tumor growth was verified using the xenograft tumor model in vivo. Effects of breast cancer cells on the proliferation and apoptosis of CD8+ T cells were analyzed using flow cytometry. Interaction between RBM15 and KPNA2 was validated using methylated RNA immunoprecipitation (MeRIP) and dual-luciferase reporter assays. RESULTS RBM15 and KPNA2 were highly expressed in breast cancer tissues and cell lines. Furthermore, RBM15 silencing might suppress breast cancer cell proliferation, migration, invasion, and lymphocyte immunity in vitro, as well as block tumor growth in vivo. At the molecular level, RBM15 might improve the stability and expression of KPNA2 mRNA via m6A methylation. CONCLUSION RBM15 might contribute to the malignant progression and immune escape of breast cancer cells partly by modulating the stability of KPNA2 mRNA, providing a promising therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Hu Wang
- Two Ward of Breast Surgery, Cancer Hospital Affiliated to Xinjiang Medical University, Urumqi city, China
| | - Yu Cao
- One Ward of Breast Surgery, Cancer Hospital Affiliated to Xinjiang Medical University, Urumqi city, China
| | - Li Zhang
- Department of Pharmacy, Cancer Hospital Affiliated to Xinjiang Medical University, Urumqi city, China
| | - Qian Zhao
- One Ward of Breast Surgery, Cancer Hospital Affiliated to Xinjiang Medical University, Urumqi city, China
| | - Shuangjian Li
- One Ward of Breast Surgery, Cancer Hospital Affiliated to Xinjiang Medical University, Urumqi city, China
| | - Dan Li
- One Ward of Breast Surgery, Cancer Hospital Affiliated to Xinjiang Medical University, Urumqi city, China.
| |
Collapse
|
39
|
Li J, Gong C, Zhou H, Liu J, Ha W, Jiang Y, Xiong H. Characterization of Immune Landscape Based on Homologous Recombination Deficiency Associated Signatures and Identification of Knockdown of ERCC6L to Promote Radiosensitivity in Breast Cancer. J Gene Med 2025; 27:e70012. [PMID: 39988478 DOI: 10.1002/jgm.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/10/2025] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Homologous recombination deficiency (HRD) exhibits significant associations with the occurrence, progression, and prognosis of breast cancer. However, the primary breast cancer HRD positivity rate is merely 24%. The identification of markers associated with HRD is crucial for the development of novel therapeutic approaches for breast cancer. The role of the oncogene ERCC6L in breast cancer remains unclear, and its interaction with radiotherapy has yet to be explored, necessitating further investigation for clarification. METHODS We employed WGCNA to identify genes associated with the HRD score, utilizing public HRD score and genetic data from TCGA breast cancer, with their clinical characteristics. Subsequently, we employed various machine learning methods to filter relevant genes. The final four genes were obtained through random forest and stepCox, and their performance was validated in TCGA, GSE96058, and METABRIC datasets. Next, we assessed the tumor immune microenvironment using methods such as ssGSEA, GSVA, CIBERSORT, ESTIMATE, and single-cell analysis. Finally, we validated the downregulation of ERCC6L, increasing DNA damage and enhancing radiation sensitivity, through immune fluorescence, flow cytometry, plate cloning, and western blot. RESULTS A prognostic model named HRAS was established through machine learning, consisting of four genes (ERCC6L, UBE2T, TPX2, and SLC7A5). The indicator exhibited excellent predictive performance on the prognosis and the efficacy of immunotherapy and radiotherapy of breast cancer patients in independent datasets. Breast cancer patients with high HRAS scores showed higher TMB and stemness, increased expression of immune checkpoints, reduced immune cell infiltration, and poorer prognosis in the context of immunotherapy and radiotherapy. Experimental validation demonstrated that knockdown of ERCC6L markedly elevated DNA damage, enhanced apoptosis, and induced cell cycle arrest in response to radiation therapy, thereby sensitizing cells to radiation. CONCLUSION The HRD-related signatures displayed strong predictive capabilities for the prognosis in multiple datasets and the efficacy of immunotherapy and radiotherapy of breast cancer patients. Moreover, the composite indicator reflected the immune microenvironment characteristics and could be novel markers for predicting the prognosis and clinical treatment outcomes in breast cancer patients. Our experiments first elucidated the role of ERCC6L in enhancing radiation-induced DNA damage, presenting a novel target for strategies aimed at sensitizing cancer cells to radiotherapy.
Collapse
Affiliation(s)
- Jiahao Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiting Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junxia Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wentao Ha
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yizhi Jiang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Famta P, Shah S, Vambhurkar G, Pandey G, Bagasariya D, Kumar KC, Prasad SB, Shinde A, Wagh S, Srinivasarao DA, Kumar R, Khatri DK, Asthana A, Srivastava S. Amelioration of breast cancer therapies through normalization of tumor vessels and microenvironment: paradigm shift to improve drug perfusion and nanocarrier permeation. Drug Deliv Transl Res 2025; 15:389-406. [PMID: 39009931 DOI: 10.1007/s13346-024-01669-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer among women. Chemo-, immune- and photothermal therapies are employed to manage BC. However, the tumor microenvironment (TME) prevents free drugs and nanocarriers (NCs) from entering the tumor premises. Formulation scientists rely on enhanced permeation and retention (EPR) to extravasate NCs in the TME. However, recent research has demonstrated the inconsistent nature of EPR among different patients and tumor types. In addition, angiogenesis, high intra-tumor fluid pressure, desmoplasia, and high cell and extracellular matrix density resist the accumulation of NCs in the TME. In this review, we discuss TME normalization as an approach to improve the penetration of drugs and NCSs in the tumor premises. Strategies such as normalization of tumor vessels, reversal of hypoxia, alleviation of high intra-tumor pressure, and infiltration of lymphocytes for the reversal of therapy failure have been discussed in this manuscript. Strategies to promote the infiltration of anticancer immune cells in the TME after vascular normalization have been discussed. Studies strategizing time points to administer TME-normalizing agents are highlighted. Mechanistic pathways controlling the angiogenesis and normalization processes are discussed along with the studies. This review will provide greater tumor-targeting insights to the formulation scientists.
Collapse
Affiliation(s)
- Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Deepkumar Bagasariya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Kondasingh Charan Kumar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Sajja Bhanu Prasad
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Akshay Shinde
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Suraj Wagh
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
- Molecular and Cellular Biology Laboratory, Department of Pharmacology, Nims Institute of Pharmacy, Nims University, Jaipur, Rajasthan, India
| | - Amit Asthana
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
41
|
Fujimoto A, Ikeda K, Kinowaki K, Ogura T, Takeiwa T, Kawabata H, Osaki A, Horie K, Inoue S. Combined use of immunoreactivities of Efp and ZCCHC3 for predicting prognosis of patients with triple-negative breast cancer. Pathol Int 2025; 75:92-99. [PMID: 39936817 DOI: 10.1111/pin.13510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/09/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
We previously reported that strong immunoreactivity (IR) of estrogen-responsive finger protein (Efp), also known as tripartite motif-containing 25 (TRIM25), predicts poor prognosis in patients with estrogen receptor-positive and -negative invasive breast cancers. In the present study, we investigated the clinicopathological role of Efp and ZCCHC3, the latter of which is an Efp interactor, in a triple-negative breast cancer (TNBC) cohort which was composed of 118 Japanese female breast cancer patients underwent surgical treatment. Efp and ZCCHC3 IRs were analyzed using specific antibodies for these proteins. We demonstrated that positive Efp IR was significantly associated with shorter distant disease-free survival (p = 0.0108) and that positive ZCCHC3 IR was also significantly associated with shorter distant disease-free survival (p = 0.0153). Notably, ZCCHC3 IR was positively associated with Efp IR (p = 0.003). When IRs of the two proteins were combined, double positivity was associated with shorter distant disease-free survival (p = 0.0007) and was an independent factor for poor prognosis. These results suggest that IR positivity of Efp and ZCCHC3 has clinical significance as a poor prognostic factor in patients with TNBC. Thus, we propose that the combined use of both IRs can be used as a prognostic marker for TNBC.
Collapse
Affiliation(s)
- Akihiro Fujimoto
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Saitama, Japan
- Department of Breast Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kazuhiro Ikeda
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Saitama, Japan
| | | | - Takuya Ogura
- Department of Breast and Endocrine Surgery, Toranomon Hospital, Japan
| | - Toshihiko Takeiwa
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Hidetaka Kawabata
- Department of Breast and Endocrine Surgery, Toranomon Hospital, Japan
| | - Akihiko Osaki
- Department of Breast Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kuniko Horie
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Saitama, Japan
| | - Satoshi Inoue
- Division of Systems Medicine & Gene Therapy, Saitama Medical University, Saitama, Japan
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| |
Collapse
|
42
|
Novakova A, Morris SA, Vaiarelli L, Frank S. Manufacturing and Financial Evaluation of Peptide-Based Neoantigen Cancer Vaccines for Triple-Negative Breast Cancer in the United Kingdom: Opportunities and Challenges. Vaccines (Basel) 2025; 13:144. [PMID: 40006691 PMCID: PMC11860436 DOI: 10.3390/vaccines13020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
This review evaluates the financial burden of current treatments for triple-negative breast cancer (TNBC) and projects potential financial scenarios to assess the feasibility of introducing a peptide-based neoantigen cancer vaccine (NCV) targeting the disease, using the UK as a healthcare system model. TNBC, the most aggressive breast cancer subtype, is associated with poor prognosis, worsened by the lack of personalised treatment options. Neoantigen cancer vaccine therapies present a personalised alternative with the potential to enhance T-cell responses independently of genetic factors, unlike approved immunotherapies for TNBC. Through a systematic literature review, the underlying science and manufacturing processes of NCVs are explored, the direct medical costs of existing TNBC treatments are enumerated, and two contrasting pricing scenarios for NCV clinical adoption are evaluated. The findings indicate that limited immunogenicity is the main scientific barrier to NCV clinical advancement, alongside production inefficiencies. Financial analysis shows that the UK spends approximately GBP 230 million annually on TNBC treatments, ranging from GBP 2200 to GBP 54,000 per patient. A best-case pricing model involving government-sponsored NCV therapy appears financially viable, while a worst-case, privately funded model exceeds the National Institute for Health and Care Excellence (NICE) cost thresholds. This study concludes that while NCVs show potential clinical benefits for TNBC, uncertainties about their standalone efficacy make their widespread adoption in the UK unlikely without further clinical research.
Collapse
Affiliation(s)
| | | | - Ludovica Vaiarelli
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| | - Stefanie Frank
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| |
Collapse
|
43
|
Liu M, Zheng L, Zhang Y, Tian J. Mechanistic insights into pachymic acid's action on triple-negative breast Cancer through TOP2A targeting. Sci Rep 2025; 15:2856. [PMID: 39843552 PMCID: PMC11754797 DOI: 10.1038/s41598-025-87286-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by the absence of estrogen and progesterone receptors, and lack of human epidermal growth factor receptor 2 (HER2) expression. Traditional Chinese medicine (TCM) has demonstrated promising efficacy in treating TNBC. This study explored the mechanisms of pachymic acid (PA) on TNBC by merging network pharmacology with experimental validation. We acquired Microarray data of TNBC from the Gene Expression Omnibus (GEO). The related targets of PA were predicted and screened using the following 6 databases: Swiss Target Prediction, HERB (Herbal Medicine Database), ETCM (Encyclopedia of Traditional Chinese Medicine), BATMAN (Bioinformatics Analysis Tool for Molecular Mechanism of Traditional Chinese Medicine), HIT (Herb Ingredients' Targets Database), and PharmMapper. The STRING interaction network analysis tool was used to create Protein-Protein Interaction (PPI) networks. Enrichment analysis included Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). We conducted a pan-cancer analysis, tumor immune microenvironment analysis, and molecular docking. We performed cell experimental, included cytotoxicity assay, apoptosis analysis, proliferation assay, and migration and invasion assays. PA has potential for treating TNBC with the target of TOP2A, and platinum drug resistance possibly serving as the KEGG pathway through which PA exerts its therapeutic effects. PA is involved in processes such as nuclear division, chromosome segregation, mitotic nuclear division, condensed chromosome formation, and protein C-terminus binding. PA probably exert its therapeutic effects through the tumor immune microenvironment, involving elements such as Dendritic cells activated, Eosinophils, Macrophages M0, Macrophages M1, and T cells CD4 memory activated. The therapeutic effects of PA may vary across different subtypes of TNBC such as TNBC-BL1, TNBC-Metaplastic, and TNBC-BL2. This study provides compelling evidence that PA holds significant promise as a therapeutic agent for TNBC, primarily through its action on TOP2A and its influence on the TNBC.
Collapse
Affiliation(s)
- Ming Liu
- Evidence-based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou City, No.199 Donggang West Road, 730000, Gansu Province, China
| | - Li Zheng
- Department of Pharmacy, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| | - Yang Zhang
- Department of Traditional Chinese medicine, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| | - Jinhui Tian
- Evidence-based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou City, No.199 Donggang West Road, 730000, Gansu Province, China.
- Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou City, Gansu Province, China.
| |
Collapse
|
44
|
Cheng C, Wang Y, Zhao J, Wu D, Li H, Zhao H. Deep Learning and Radiomics in Triple-Negative Breast Cancer: Predicting Long-Term Prognosis and Clinical Outcomes. J Multidiscip Healthc 2025; 18:319-327. [PMID: 39866348 PMCID: PMC11762009 DOI: 10.2147/jmdh.s509004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/11/2025] [Indexed: 01/28/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a unique breast cancer subtype characterized by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression in tumor cells. TNBC represents about 15% to 20% of all breast cancers and is aggressive and highly malignant. Currently, TNBC diagnosis primarily depends on pathological examination, while treatment efficacy is assessed through imaging, biomarker detection, pathological evaluation, and clinical symptom improvement. Among these, biomarker detection and pathological assessments are invasive, time-intensive procedures that may be difficult for patients with severe comorbidities and high complication risks. Thus, there is an urgent need for new, supportive tools in TNBC diagnosis and treatment. Deep learning and radiomics techniques represent advanced machine learning methodologies and are also emerging outcomes in the medical-engineering field in recent years. They are extensions of conventional imaging diagnostic methods and have demonstrated tremendous potential in image segmentation, reconstruction, recognition, and classification. These techniques hold certain application prospects for the diagnosis of TNBC, assessment of treatment response, and long-term prognosis prediction. This article reviews recent progress in the application of deep learning, ultrasound, MRI, and radiomics for TNBC diagnosis and treatment, based on research from both domestic and international scholars.
Collapse
Affiliation(s)
- Chen Cheng
- Department of Ultrasound, Lianyungang Traditional Chinese Medicine Hospital, Lianyungang, 222004, People’s Republic of China
| | - Yan Wang
- Department of Ultrasound, Lianyungang Municipal Oriental Hospital, Lianyungang, 222046, People’s Republic of China
- Department of Ultrasound, Xuzhou Medical University Affiliated Hospital, Lianyungang, Jiangsu, 222061, People’s Republic of China
| | - Jine Zhao
- Department of Ultrasound, Donghai County People’s Hospital, Lianyungang, Jiangsu, 222300, People’s Republic of China
| | - Di Wu
- Department of Ultrasound, Lianyungang Traditional Chinese Medicine Hospital, Lianyungang, 222004, People’s Republic of China
| | - Honge Li
- Department of Ultrasound, the First People’s Hospital of Lianyungang, Lianyungang, Jiangsu, 222061, People’s Republic of China
| | - Hongyan Zhao
- Department of Ultrasound, Lianyungang Traditional Chinese Medicine Hospital, Lianyungang, 222004, People’s Republic of China
| |
Collapse
|
45
|
Chen Z, Zhao Y. The mechanism underlying metastasis in triple-negative breast cancer: focusing on the interplay between ferroptosis, epithelial-mesenchymal transition, and non-coding RNAs. Front Pharmacol 2025; 15:1437022. [PMID: 39881868 PMCID: PMC11774878 DOI: 10.3389/fphar.2024.1437022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a type of breast cancer with lack the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). It is the most aggressive breast cancer and the most difficult to treat due to its poor response to treatments and extremely invasive characteristics. The typical treatment for TNBC frequently results in relapse because of the lack of particular treatment choices. It is urgent to focus on identifying a workable and effective target for the treatment of TNBC. Cancer metastasis is significantly influenced by epithelial-mesenchymal transition (EMT). Ferroptosis is an iron-dependent cell death form, and changes its key factor to affect the proliferation and metastasis of TNBC. Several reports have established associations between EMT and ferroptosis in TNBC metastasis. Furthermore, non-coding RNA (ncRNA), which has been previously described, can also control cancer cell death and metastasis. Thus, in this review, we summarize the correlation and pathways among the ferroptosis, EMT, and ncRNAs in TNBC metastasis. Also, aim to find out a novel strategy for TNBC treatment through the ncRNA-ferroptosis-EMT axis.
Collapse
Affiliation(s)
- Ziyi Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yi Zhao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Healthcare Hospital of Shandong Province Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
46
|
Khalaji A, Hadad S, Jabbaripour Sarmadian A, Ojaghlou F, Janagard EM, Baradaran B. Advances in Monoclonal Antibody Therapies for Triple-Negative Breast Cancer: Immunotherapeutic and Targeted Strategies. Curr Mol Med 2025; 25:445-459. [PMID: 38288828 DOI: 10.2174/0115665240287767240115062343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 02/07/2024]
Abstract
Triple-negative breast cancer (TNBC) presents considerable obstacles because of its highly aggressive characteristics and limited availability of specific therapeutic interventions. The utilization of monoclonal antibody (mAb)-based immunotherapy is a viable approach to tackle these difficulties. This review aims to examine the present state of mAb-based immunotherapy in TNBC, focusing on the underlying mechanisms of action, clinical applications, and existing challenges. The effectiveness of mAbs in reducing tumor development, regulating immune responses, and changing the tumor microenvironment has been demonstrated in many clinical investigations. The challenges encompass several aspects such as the discovery of biomarkers, understanding resistance mechanisms, managing toxicity, considering costs, and ensuring accessibility. The future is poised to bring forth significant advancements in the field of biomedicine, particularly in the areas of new mAbs, personalized medicine, and precision immunotherapy. In conclusion, mAb-based immunotherapy has promise in revolutionizing the treatment of TNBC, hence providing a possible avenue for enhanced patient outcomes and quality of life.
Collapse
Affiliation(s)
- Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Hadad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirreza Jabbaripour Sarmadian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ojaghlou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
47
|
Peng Y, Qi X, Ding L, Huang J, Liu Y, Zheng R, Fu Y, Yin L, Deng T, Ye Y, Chen S, Li X. SKP2 inhibition activates tumor cell-intrinsic immunity by inducing DNA replication stress and genomic instability. Br J Cancer 2025; 132:81-92. [PMID: 39582087 PMCID: PMC11723935 DOI: 10.1038/s41416-024-02909-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND S-phase kinase-associated protein 2 (SKP2) is a typical oncogene aberrantly overexpressing in a variety of cancer types, but it remains elusive whether SKP2 regulates the antitumor immunity of triple-negative breast cancer. METHODS The efficacy of anti-PD-1 was evaluated in the orthotopic xenografts of immunocompetent mice models. The infiltration of cytotoxic T cells in tumor microenvironment(TME) were assessed by immunofluorescence staining. The levels of pro-inflammatory chemokines were analyzed by ELISA. The protein interaction was analyzed by co-immunoprecipitation and GST pull-down. The genomic instability was analyzed by fluorescent microscopy. RESULTS SKP2 inhibition significantly improved the antitumor efficacy of immune checkpoint blockade (ICB). Furthermore, SKP2 inhibition activated the cGAS/STING signal pathway and induced the secretion of pro-inflammatory chemokines, thereby promoting cytotoxic T cell infiltration. Additionally, we identified CDC6, a DNA replication licensing factor as a novel substrate of SKP2 in addition to CDT1. SKP2 induced protein degradation of CDC6 and CDT1 through the ubiquitin-proteasome pathway. Conversely, SKP2 inhibition elevated CDC6 and CDT1 protein levels, which caused DNA aberrant replication, DNA damage and genomic instability, thereby resulting in the accumulation of cytosolic DNA, activating cGAS/STING signaling pathway and improving antitumor immunity. CONCLUSION SKP2 may be used as an effective therapeutic target to enable ICB antitumor immunotherapy. SOCIAL MEDIA Peng et al. found that SKP2 inhibition improved the antitumor immunotherapy by activating tumor cell-intrinsic immunity, thereby providing evidences that SKP2 may be used as an effective therapeutic target to enable ICB antitumor immunotherapy.
Collapse
Affiliation(s)
- Yuchong Peng
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
- Institute of Integrative Neurology of integrated traditional Chinese and Western Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
| | - Xuli Qi
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Liuyang Ding
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Jingjing Huang
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Youhong Liu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Rirong Zheng
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Yongming Fu
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
- Institute of Integrative Neurology of integrated traditional Chinese and Western Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
| | - Linglong Yin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Tanggang Deng
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Yubing Ye
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Size Chen
- Department of Tumor Immunity, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China
| | - Xiong Li
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China.
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510699, China.
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China.
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
48
|
Xu Y, Cai Y, Deng Y, He Y, Wu J, Chang S, Yan X, Wang J. RAC2 as a Tumor-Suppressive Biomarker Associated with T Cell Infiltration in Breast Cancer. Cancer Biother Radiopharm 2025; 40:62-77. [PMID: 39479793 DOI: 10.1089/cbr.2024.0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025] Open
Abstract
Background: RAC2 is critical in regulating the homeostasis of hematopoietic stem cells. Nonetheless, its role in breast cancer (BC) remains unclear, necessitating further investigation. Methods: The expression of RAC2 in BC and healthy tissues was acquired from The Cancer Genome Atlas. Its validity was further assessed using datasets from the gene expression omnibus database. The Tumor Immune Single-cell Hub database was used to collect and analyze the single-cell RNA sequencing datasets of BC. The diagnostic relevance of RAC2 was evaluated using receiver operating characteristic curves. Further assessment was carried out via enrichment analyses; Gene Set Analysis, immune scoring, single-cell sequencing, and immunohistochemical analysis were conducted to confirm the relationship between RAC2 expression and immune infiltration. Results: RAC2 expression was notably heightened in BC (p < 0.001). It was observed that a better prognosis was linked to heightened expression of RAC2 (p < 0.01), with the diagnostic efficacy of the marker noted to be good (area under the curve = 0.858). We found a lower percentage of protumor immune cells and a greater proportion of antitumor immune cells in the high RAC2. Our analysis revealed alterations in gene expression and an enriched network of immune pathways influenced by RAC2. Notably, cytotoxic genes, chemokines, chemokine receptors, immunostimulators, and immunosuppressive molecules positively correlated with RAC2 expression. RAC2 expression reliably predicted how patients would respond to two different therapeutic approaches in BC. Conclusions: The RAC2 was found to be a key biomarker in BC in the current study, demonstrating considerable potential as a prognostic and diagnostic marker. These results highlight the RAC2 potential to improve precision medicine strategies and treatment outcomes for patients with BC.
Collapse
Affiliation(s)
- Yiping Xu
- Department of General Surgery, Xiangtan Central Hospital (The affiliated hospital of Hunan University), Xiangtan, China
| | - Yurong Cai
- Department of General Surgery, Xiangtan Central Hospital (The affiliated hospital of Hunan University), Xiangtan, China
| | - Youyuan Deng
- Department of General Surgery, Xiangtan Central Hospital (The affiliated hospital of Hunan University), Xiangtan, China
| | - Ye He
- Department of General Surgery, Xiangtan Central Hospital (The affiliated hospital of Hunan University), Xiangtan, China
| | - Juan Wu
- Department of Pathology, Xiangtan Central Hospital, Xiangtan, China
| | - Shunqiu Chang
- Department of Pathology, Xiangtan Central Hospital, Xiangtan, China
| | - Xuebo Yan
- Department of General Surgery, Shaoyang Central Hospital, Shaoyang, China
| | - Jianguo Wang
- Department of General Surgery, Xiangtan Central Hospital (The affiliated hospital of Hunan University), Xiangtan, China
| |
Collapse
|
49
|
Chu M, Huang J, Wang Q, Fang Y, Cui D, Jin Y. A Circadian Rhythm-related Signature to Predict Prognosis, Immune Infiltration, and Drug Response in Breast Cancer. Curr Med Chem 2025; 32:608-626. [PMID: 39279697 DOI: 10.2174/0109298673320179240803071001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 09/18/2024]
Abstract
PURPOSE Circadian rhythm-related genes (CRRGs) play essential roles in cancer occurrence and development. However, the prognostic significance of CRRGs in breast cancer (BC) has not been fully elucidated. Our study aimed to develop a prognostic gene signature based on CRRGs that can accurately and stably predict the prognosis of BC. METHODS The transcriptome data and clinical information for BC patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. A consensus unsupervised clustering analysis was carried out to investigate the roles of CRRGs in BC. A CRRGs-related prognostic risk model was established by using logistic least absolute shrinkage and selection operator (LASSO) Cox regression and univariate Cox regression analyses. Kaplan-Meier (KM) curves analysis, time-dependent receptor operation characteristics (ROC) curves analysis, and nomogram were plotted to evaluate the predictive efficacy of the model. The relevance of risk score to the immune cell infiltration, tumor burden mutation (TMB), and therapeutic response was assessed. RESULTS A risk model comprising six CRRGs (SLC44A4, SLC16A6, TPRG1, FABP7, GLYATL2, and FDCSP) was constructed and validated, demonstrating an effective predictor for the prognosis of BC. The low-risk group displayed a higher expression of immune checkpoint genes and a lower burden of tumor mutation. Additionally, drug sensitivity analysis demonstrated that the prognostic signature may serve as a potential chemosensitivity predictor. CONCLUSION We established a CRRGs-related risk signature, which is of great value in predicting the prognosis of patients with BC and guiding the treatment for BC.
Collapse
Affiliation(s)
- Mingyu Chu
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Jing Huang
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Qianyu Wang
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yaqun Fang
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Dina Cui
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| | - Yucui Jin
- Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, China
| |
Collapse
|
50
|
Guo Z, Zhu Z, Lin X, Wang S, Wen Y, Wang L, Zhi L, Zhou J. Tumor microenvironment and immunotherapy for triple-negative breast cancer. Biomark Res 2024; 12:166. [PMID: 39741315 DOI: 10.1186/s40364-024-00714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer known for its high aggressiveness and poor prognosis. Conventional treatment of TNBC is challenging due to its heterogeneity and lack of clear targets. Recent advancements in immunotherapy have shown promise in treating TNBC, with immune checkpoint therapy playing a significant role in comprehensive treatment plans. The tumor microenvironment (TME), comprising immune cells, stromal cells, and various cytokines, plays a crucial role in TNBC progression and response to immunotherapy. The high presence of tumor-infiltrating lymphocytes and immune checkpoint proteins in TNBC indicates the potential of immunotherapeutic strategies. However, the complexity of the TME, while offering therapeutic targets, requires further exploration of its multiple roles in immunotherapy. In this review, we discuss the interaction mechanism between TME and TNBC immunotherapy based on the characteristics and composition of TME, and elaborate on and analyze the effect of TME on immunotherapy, the potential of TME as an immune target, and the ability of TME as a biomarker. Understanding these dynamics will offer new insights for enhancing therapeutic approaches and investigating stratification and prognostic markers for TNBC patients.
Collapse
Affiliation(s)
- Zijie Guo
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Ziyu Zhu
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Xixi Lin
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Shenkangle Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yihong Wen
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Linbo Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Lili Zhi
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|